EP2334687B9 - Dioxa-bicyclo[3.2.1.]octane-2,3,4-triol derivatives - Google Patents

Dioxa-bicyclo[3.2.1.]octane-2,3,4-triol derivatives Download PDF

Info

Publication number
EP2334687B9
EP2334687B9 EP09786960A EP09786960A EP2334687B9 EP 2334687 B9 EP2334687 B9 EP 2334687B9 EP 09786960 A EP09786960 A EP 09786960A EP 09786960 A EP09786960 A EP 09786960A EP 2334687 B9 EP2334687 B9 EP 2334687B9
Authority
EP
European Patent Office
Prior art keywords
benzyl
dioxa
bicyclo
hydroxymethyl
phenyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP09786960A
Other languages
German (de)
French (fr)
Other versions
EP2334687A1 (en
EP2334687B1 (en
Inventor
Vincent Mascitti
Benjamin Micah Collman
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pfizer Inc
Original Assignee
Pfizer Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=41208284&utm_source=***_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP2334687(B9) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Pfizer Inc filed Critical Pfizer Inc
Priority to PL09786960T priority Critical patent/PL2334687T3/en
Priority to MEP-2011-45A priority patent/ME01285A/en
Publication of EP2334687A1 publication Critical patent/EP2334687A1/en
Application granted granted Critical
Publication of EP2334687B1 publication Critical patent/EP2334687B1/en
Priority to CY20121100280T priority patent/CY1112497T1/en
Publication of EP2334687B9 publication Critical patent/EP2334687B9/en
Priority to HUS1800031C priority patent/HUS1800031I1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H9/00Compounds containing a hetero ring sharing at least two hetero atoms with a saccharide radical
    • C07H9/02Compounds containing a hetero ring sharing at least two hetero atoms with a saccharide radical the hetero ring containing only oxygen as ring hetero atoms
    • C07H9/04Cyclic acetals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/02Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains two hetero rings
    • C07D493/08Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/357Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having two or more oxygen atoms in the same ring, e.g. crown ethers, guanadrel
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers

Definitions

  • the present invention relates to dioxa-bicyclo[3.2.1]octane-2,3,4-triol derivatives, crystal structures, pharmaceutical compositions and the uses thereof as sodium-glucose co-transporter (SGLT) inhibitors.
  • SGLT sodium-glucose co-transporter
  • Obesity is a significant health problem due to its serious medical complications that include co-morbidities such as hypertension, insulin resistance, diabetes, coronary artery disease and heart failure (collectively referred to as Metabolic Syndrome). Obesity and its related co-morbidities continue to cause rising health issues in the developed world and are beginning to affect the developing world as well. The negative health consequences of obesity make it the second leading cause of preventable death in the United States and impart a significant economic and psychosocial effect on society. See, McGinnis M, Foege WH., "Actual Causes of Death in the United States," JAMA, 270, 2207-12 (1993 ). There is a need to identify and develop new medications that treat and/or prevent obesity and its associated co-morbidities, in particular type II (type 2) diabetes.
  • type II type 2
  • SGLT sodium-glucose co-transport
  • SGLT2 inhibitors sodium-glucose co-transport
  • SGLT2 inhibitiors provide an attractive means for the improvement of diabetic conditions without increasing body weight or the risk of hypoglycemia.
  • glycosides that have been shown to be useful for the treatment of NIDDM and obesity can be found in the following disclosures: U.S. Patent Nos. 6,515,117 ; 6,414,126 ; 7,101,856 ; 7,169,761 ; and 7,202,350 ; U.S. Publication Nos. US2002/0111315 ; US2002/0137903 ; US2004/0138439 ; US2005/0233988 ; US2006/0025349 ; US2006/0035841 ; and US2006/0632722 ; and PCT Publication Nos.
  • Certain glycosides are genotoxic and impact a cell's genetic material such that they may be potentially mutagenic or carcinogenic. Genotoxic materials may be detected using standard assays such as the In Vitro Mammalian Cell Micronuleus Test (MNvit), Organization for Economic Co-Operation and Development (OECD) Draft Test Guideline (Draft TG) 487 (2007); In vitro Mammalian Chromosomal Aberration Test, OECD TG 473 (1997); Bacterial Reverse Mutation Test, OECD TG 471 (1997); Mammalian Erythrocyte Micronucleus Test, OECD TG 474 (1997); or the like. Consequently, there still exists a need for a more effective and safe therapeutic treatment and/or prevention of obesity and its associated co-morbidities, in particular, Type 2 diabetes and related disorders.
  • MNvit In Vitro Mammalian Cell Micronuleus Test
  • OECD Organization for Economic Co-Operation and Development
  • SGLT sodium-glucose cotransport
  • compounds of Formula (A) and Formula (B) have been found to act as sodium-glucose cotransport (SGLT) inhibitors, in particular, SGLT2 inhibitors; therefore, may be used in the treatment of diseases mediated by such inhibition (e.g., diseases related to obesity, Type 2 diabetes, and obesity-related and diabetes-related co-morbidities).
  • diseases mediated by such inhibition e.g., diseases related to obesity, Type 2 diabetes, and obesity-related and diabetes-related co-morbidities.
  • R 1 is H, (C 1 -C 4 )alkyl, (C 1 -C 4 )alkoxy, Cl, F, cyano, fluoro-substituted (C 1 -C 2 )alkyl, (C 1 -C 4 )alkyl-SO 2 -, or (C 3 -C 6 )cycloalkyl; and R 2 is (C 1 -C 4 )alkyl, (C 1 -C 4 )alkoxy, (C 2 -C 4 )alkynyl, 3-oxetanyloxy, 3-tetrahydrofuranyloxy, Cl, F, cyano, fluoro-substituted (C 1 -C 2 )alkyl, (C 1 -C 4 )alkyl-SO 2 -, (C 3 -C 6 )cycloalkyl, or a (C 5 -C 6 )
  • Specific compounds of Formula (A) include: (1S,2S,3S,4R,5S)-1-hydroxymethyl-5-[3-(4-methoxy-benzyl)-4-methyl-phenyl]-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4R,5S)-5-[3-(4-ethoxy-benzyl)-4-methyl-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4R,5S)-5-[4-chloro-3
  • Specific compounds of Formula (B) include: (1S,2S,3S,4S,5S)-1-hydroxymethyl-5-[3-(4-methoxy-benzyl)-4-methyl-phenyl]-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4S,5S)-5-[3-(4-ethoxy-benzyl)-4-methyl-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4S,5S)-5-[4-chloro-3-(4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4S,5S)-5-[4-chloro-3
  • a further aspect of the present invention is a crystal comprising a compound having the formula (4A):
  • compositions that comprises (1) a compound of the present invention, and (2) a pharmaceutically acceptable excipient, diluent, or carrier.
  • the composition comprises a therapeutically effective amount of a compound of the present invention.
  • the composition may also contain at least one additional pharmaceutical agent (described herein).
  • Preferred agents include anti-obesity agents and/or anti-diabetic agents (described herein below).
  • a use of the compounds of the present invention in a method for treating a disease, disorder, or condition modulated by SGLT2 inhibition in animals includes the step of administering to an animal (preferably, a human) in need of such treatment a therapeutically effective amount of a compound of the present invention (or a pharmaceutical composition thereof).
  • Diseases, conditions, and/or disorders modulated by SGLT2 inhibition include, e.g., Type II diabetes, diabetic nephropathy, insulin resistance syndrome, hyperglycemia, hyperinsulinemia, hyperlipidemia, impaired glucose tolerance, obesity (including weight control or weight maintenance), hypertension, and reducing the level of blood glucose.
  • Compounds of the present invention may be administered in combination with other pharmaceutical agents (in particular, anti-obesity and anti-diabetic agents described herein below).
  • the combination therapy may be administered as (a) a single pharmaceutical composition which comprises a compound of the present invention, at least one additional pharmaceutical agent described herein and a pharmaceutically acceptable excipient, diluent, or carrier; or (b) two separate pharmaceutical compositions comprising (i) a first composition comprising a compound of the present invention and a pharmaceutically acceptable excipient, diluent, or carrier, and (ii) a second composition comprising at least one additional pharmaceutical agent described herein and a pharmaceutically acceptable excipient, diluent, or carrier.
  • the pharmaceutical compositions may be administered simultaneously or sequentially and in any order.
  • cycloalkyl refers to nonaromatic rings that are fully hydrogenated and may exist as a single ring, bicyclic ring or a spiro ring. Unless specified otherwise, the carbocyclic ring is generally a 3- to 8-membered ring.
  • cycloalkyl include groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, norbornyl (bicyclo[2.2.1]heptyl), bicyclo[2.2.2]octyl, and the like.
  • heterocycle refers to nonaromatic rings that are fully hydrogenated and may exist as a single ring, bicyclic ring or a spiral ring. Unless specified otherwise, the heterocyclic ring is generally a 3- to 6-membered ring containing 1 to 3 heteroatoms (preferably 1 or 2 heteroatoms) independently selected from sulfur, oxygen and/or nitrogen.
  • Heterocyclic rings include groups such as epoxy, aziridinyl, tetrahydrofuranyl, pyrrolidinyl, N-methylpyrrolidinyl, piperidinyl, piperazinyl, pyrazolidinyl, 4H-pyranyl, morpholino, thiomorpholino, tetrahydrothienyl, tetrahydrothienyl 1,1-dioxide, and the like.
  • terapéuticaally effective amount means an amount of a compound of the present invention that (i) treats the particular disease, condition, or disorder, (ii) attenuates, ameliorates, or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein.
  • animal refers to humans (male or female), companion animals (e.g., dogs, cats and horses), food-source animals, zoo animals, marine animals, birds and other similar animal species.
  • companion animals e.g., dogs, cats and horses
  • food-source animals e.g., zoo animals, marine animals, birds and other similar animal species.
  • Edible animals refers to food-source animals such as cows, pigs, sheep and poultry.
  • phrases "pharmaceutically acceptable” indicates that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
  • treating embrace both preventative, i.e., prophylactic, and palliative treatment.
  • modulated refers to the inhibition of the sodium-glucose transporter (in particular, SGLT2) with compounds of the present invention thereby partially or fully preventing glucose transport across the transporter.
  • compounds of the present invention refer to compounds of Formula (A), Formula (B) and all pure and mixed stereoisomers (including diastereoisomers and enantiomers), tautomers and isotopically labeled compounds. Hydrates and solvates of the compounds of the present invention are considered compositions of the present invention, wherein the compound is in association with water or solvent, respectively.
  • the compounds may also exist in one or more crystalline states, i.e. as co-crystals, polymorphs, or they may exist as amorphous solids. All such forms are encompassed by the claims.
  • R 1 is H, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, F, Cl, cyano, -CF 3 , cyclopropyl, or cyclobutyl.
  • R 1 is H, methyl, ethyl, isopropyl, methoxy, ethoxy, F, Cl, cyano, -CF 3 , or cyclopropyl.
  • R 1 is H, methyl, ethyl, methoxy, ethoxy, F, Cl, cyano, -CF 3 , or cyclopropyl.
  • R 1 is methyl, ethyl, F, Cl, cyano, CF 3 , or cyclopropyl.
  • R 2 is methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, F, Cl, cyano, -CF 3 , -CF 2 CH 3 , ethynyl, 3-oxetanyloxy, 3-tetrahydrofuranyloxy, or cyclopropyl.
  • R 2 is methyl, ethyl, isopropyl, methoxy, ethoxy, F, Cl, cyano, -CF 3 ,-CF 2 CH 3 , ethynyl, 3-oxetanyloxy, 3-tetrahydrofuranyloxy, or cyclopropyl.
  • R 2 is methyl, ethyl, methoxy, ethoxy, F, Cl, cyano, -CF 3 , -CF 2 CH 3 , ethynyl, 3-oxetanyloxy, 3-tetrahydrofuranyloxy, or cyclopropyl.
  • R 2 is methoxy or ethoxy.
  • the crystal comprises the compound 4A and L-proline or L-pyroglutamic acid.
  • the crystal has one or more of the following:
  • the crystal is a co-crystal comprising the compound of formula (4A) and L-pyroglutamic acid in a 1:1 stochiometric ratio.
  • Compounds of the present invention may be synthesized by synthetic routes that include processes analogous to those well-known in the chemical arts, particularly in light of the description contained herein.
  • the starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, WI) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v. 1-19, Wiley, New York (1967-1999 ed .), or Beilsteins Handbuch der organischen Chemie, 4, Aufl. ed. Springer-Verlag, Berl in, including supplements (also available via the Beilstein online database)).
  • reaction schemes depicted below provide potential routes for synthesizing the compounds of the present invention as well as key intermediates.
  • Examples section below For a more detailed description of the individual reaction steps, see the Examples section below.
  • Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the inventive compounds.
  • specific starting materials and reagents are depicted in the schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions.
  • many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
  • hydroxy-protecting group refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality.
  • Suitable hydroxyl-protecting groups include for example, allyl, acetyl (Ac), silyl (like trimethylsily (TMS) or tert-butyldimethylsilyl (TBS)), benzyl (Bn), para -methoxybenzyl (PMB), trityl (Tr), para -bromobenzoyl, para- nitrobenzoyl and the like (benzylidene for protection of 1,3-diols).
  • TMS trimethylsily
  • TSS tert-butyldimethylsilyl
  • PMB para -methoxybenzyl
  • Tr trityl
  • para -bromobenzoyl para-nitrobenzoyl and the like
  • Scheme 1 outlines the general procedures one could use to provide compounds of the present invention.
  • Allyl 2,3,4-tri-O-benzyl-D-glucopyranoside ( I-a , where Pg 1 is a benzyl group) can be prepared by procedures described by Shinya Hanashima, et al., in Bioorganic & Medicinal Chemistry, 9, 367 (2001 ); Patricia A. Gent et al. in Journal of the Chemical Society. Perkin 1, 1835 (1974 ); Hans Peter Wessel in the Journal of Carbohydrate Chemistry, 7, 263, (1988 ); or Yoko Yuasa, et al., in Organic Process Research & Development, 8, 405-407 (2004 ).
  • the hydroxymethylene group can be introduced onto the glycoside by means of a Swern oxidation followed by treatment with formaldehyde in the presence of an alkali metal hydroxide (e.g., sodium hydroxide).
  • an alkali metal hydroxide e.g., sodium hydroxide
  • the Swern oxidation is described by Kanji Omura and Daniel Swern in Tetrahedron, 34, 1651 (1978 ). Modifications of this process known to those of skill in the art may also be used.
  • other oxidants like stabilized 2-iodoxybenzoic acid described by Ozanne, A. et al.
  • protecting groups (Pg 2 ) can be added by treating intermediate ( I-b ) with the appropriate reagents and procedures for the particular protecting group desired.
  • PMB p -methoxybenzyl
  • protecting groups (Pg 2 ) can be added by treating intermediate ( I-b ) with the appropriate reagents and procedures for the particular protecting group desired.
  • p -methoxybenzyl (PMB) groups may be introduced by treatment of intermediate ( I-b ) with p-methoxybenzyl bromide or p -methoxybenzyl chloride in the presence of sodium hydride, potassium hydride, potassium tert -butoxide in a solvent like tetrahydrofuran, 1,2-dimethoxyethane or N , N -dimethylformamide (DMF).
  • Conditions involving para-methoxybenzyltrichloroacetimidate in presence of a catalytic amount of acid (e.g., trifluoromethanesulfonic acid, methanesulfonic acid, or camphorsulfonic acid) in a solvent such as dichloromethane, heptane or hexanes can also be used.
  • Benzyl (Bn) groups may be introduced by treatment of intermediate ( I-b ) with benzyl bromide or benzyl chloride in the presence of sodium hydride, potassium hydride, potassium tert -butoxide in a solvent like tetrahydrofuran, 1,2-dimethoxyethane or N , N -dimethylformamide.
  • Conditions involving benzyltrichloroacetimidate in presence of a catalytic amount of acid e.g., trifluoromethanesulfonic acid, methanesulfonic acid, or camphorsulfonic acid
  • a catalytic amount of acid e.g., trifluoromethanesulfonic acid, methanesulfonic acid, or camphorsulfonic acid
  • a solvent such as dichloromethane, heptane or hexanes
  • step 3 of Scheme 1 the allyl protection group is removed (e.g., by treatment with palladium chloride in methanol; cosolvent like dichloromethane may also be used; other conditions known by those skilled in the art could also be used, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991 ) to form the lactol ( I-d ).
  • step 4 of Scheme 1 oxidation of the unprotected hydroxyl group to an oxo group (e.g., Swern oxidation) then forms the lactone ( I-e ).
  • an oxo group e.g., Swern oxidation
  • step 5 of Scheme 1 the lactone (I-e ) is reacted with N , O -dimethyl hydroxylamine hydrochloride to form the corresponding Weinreb amide which may exist in equilibrium in a closed/opened form.
  • the "Weinreb amide" ( I-g ) can be made using procedures well known to those of skill in the art. See, Nahm, S., and S.M. Weinreb, Tetrahedron Letters, 22 (39), 3815-1818 (1981 ).
  • intermediate (I-f/I-g ) can be prepared from the commercially available N , O -dimethylhydroxylamine hydrochloride and an activating agent (e.g., trimethylaluminum).
  • the arylbenzyl group (Ar) is introduced using the desired organometallic reagent (e.g., organo lithium compound (ArLi) or organomagnesium compound (ArMgX)) in tetrahydrofuran (THF) at a temperature ranging from about -78°C to about 20°C followed by hydrolysis (upon standing in protic conditions) to the corresponding lactol ( I-i ) which may be in equilibrium with the corresponding ketone ( I-h ).
  • the bridged ketal motif found in (A) and (B) can be prepared by removing the protecting groups (Pg 2 ) using the appropriate reagents for the protecting groups employed.
  • the PMB protecting groups may be removed by treatment with trifluoroacetic acid in the presence of anisole and dichloromethane (DCM) at about 0°C to about 23°C (room temperature).
  • DCM anisole and dichloromethane
  • the remaining protecting groups (Pg 1 ) may then be removed using the appropriate chemistry for the particular protecting groups.
  • benzyl protecting groups may be removed by treating with formic acid in the presence of palladium (Pd black) in a protic solvent (e.g., ethanol/THF) at about room temperature to produce the final products (A) and (B).
  • a protic solvent e.g., ethanol/THF
  • R 1 is CN
  • a Lewis acid like boron trichloride at a temperature ranging from about -78°C to about room temperature in a solvent like dichloromethane or 1,2-dichloroethane may also be used to remove benzyl protective and/or para-methoxybenzyl protective groups.
  • R 1 is CN and R 2 is (C 1 -C 4 )alkoxy in intermdediate (I-i) or in products (A) or (B)
  • a Lewis acid such as boron trichloride or boron tribomide
  • partial to complete de-alkylation to the corresponding phenol may occur to lead to the corresponding compound (A) or (B) where R 1 is CN and R 2 is OH.
  • the (C 1 -C 4 )alkoxy group may be re-introduced via selective alkylation using a (C 1 -C 4 ) alkyl iodide under mildly basic conditions, for example, potassium carbonate in acetone at a temperature ranging from about room temperature to about 56 degrees Celsius.
  • R 1 and/or R 2 is (C 1 -C 4 )alkyl-SO 2 - it is understood by one skilled in the art that the organometallic addition step 6 (Scheme 1) will be carried out on the corresponding (C 1 -C 4 )alkyl-S- containing organometallic reagent.
  • the thio-alkyl is then oxidized at a later stage to the corresponding sulfone using conventional methods known by those skilled in the art.
  • the compounds of the present invention may be prepared as co-crystals using any suitable method.
  • a representative scheme for preparing such co-crystals is described in Scheme 2.
  • step 2 of Scheme 2 to (II-b) in methylene chloride is added imidazole followed by cooling to 0°C and then addition of trimethylsilylchloride to give the persilylated product. The reaction is warmed to room temperature and quenched by the addition of water, and the organic phase is washed with water. This crude methylene chloride solution of (II-c) is dried over sodium sulfate and then taken on crude into the next step.
  • step 3 of Scheme 2 the crude solution of (II-c) in methylene chloride is concentrated to low volume and then the solvent is exchanged to methanol.
  • the methanol solution of (II-c) is cooled to 0°C, then 1 mol% of potassium carbonate is added as a solution in methanol followed by stirring for 5 hours.
  • the reaction is then quenched by addition of 1 mol% acetic acid in methanol, followed by warming to room temperature, solvent exchange to ethyl acetate, and then filtration of the minor amount of inorganic solids.
  • the crude ethyl acetate solution of (II-d) is taken directly into the next step.
  • step 4 of Scheme 2 the crude solution of (II-d) is concentrated to low volume, then diluted with methylene chloride and dimethylsulfoxide. Triethylamine is added followed by cooling to 10°C and then sulfur trioxide pyridine complex is added in 3 portions as a solid at 10 minute intervals. The reaction is stirred an additional 3 hours at 10°C before quenching with water and warming to room temperature. The phases are separated followed by washing the methylene chloride layer with aqueous ammonium chloride. The crude methylene chloride solution of (II-e) is taken directly into the next step.
  • step 5 of Scheme 2 the crude solution of (II-e) is concentrated to low volume and then the solvent is exchanged to ethanol. Thirty equivalents of aqueous formaldehyde is added followed by warming to 55°C. An aqueous solution of 2 equivalents of potassium phosphate, tribasic is added followed by stirring for 24 hours at 55°C. The reaction temperature is then raised to 70°C for an additional 12 hours. The reaction is cooled to room temperature, diluted with tert-butyl methyl ether and brine. The phases are separated followed by solvent exchange of the organic phase to ethyl acetate. The ethyl acetate phase is washed with brine and concentrated to low volume.
  • step 6 of Scheme 2 compound (II-f) is dissolved in 5 volumes of methylene chloride followed by the addition of 1mol% Silia Bond ® tosic acid and stirring for 18 hours at room temperature. The acid catalyst is filtered off and the methylene chloride solution of (II-g) is taken directly into the next step co-crystallization procedure.
  • step 7 of Scheme 2 the methylene chloride solution of (II-g) is concentrated and then the solvent is exchanged to 2-propanol. Water is added followed by warming to 55°C. An aqueous solution of L-pyroglutamic acid is added followed by cooling the resulting solution to room temperature. The solution is then seeded and granulated for 18 hours. After cooling, the solids are collected and rinsed with heptane followed by drying. Product (II-h) is isolated as a solid.
  • step 1 of Scheme 3 the primary hydroxyl group is selectively protected by an appropriate protective group.
  • Additional examples of such protective groups and experimental conditions are known by those skilled in the art and can be found in T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991 .
  • the secondary hydroxyl groups can be protected by the appropriate protecting groups.
  • benzyl groups Pg 4 is Bn
  • benzyl groups can be introduced by treatment of intermediate (III-b) with benzyl bromide or benzyl chloride in the presence of sodium hydride, potassium hydride, potassium tert-butoxide in a solvent like tetrahydrofuran, 1,2-dimethoxyethane or N , N -dimethylformamide at a temperature ranging from about 0 degrees Celsius to about 80 degrees Celsius.
  • Acetyl or benzoyl groups may be introduced by treatment of intermediate (III-b) with acetyl chloride, acetyl bromide or acetic anhydride or benzoyl chloride or benzoic anhydride in the presence of a base like triethylamine, N , N -diisopropylethylamine or 4-(dimethylamino)pyridine in a solvent like tetrahydrofuran, 1,2-dimethoxyethane or dichloromethane at a temperature ranging from about 0 degrees Celsius to about 80 degrees Celsius.
  • a base like triethylamine, N , N -diisopropylethylamine or 4-(dimethylamino)pyridine in a solvent like tetrahydrofuran, 1,2-dimethoxyethane or dichloromethane at a temperature ranging from about 0 degrees Celsius to about 80 degrees Celsius.
  • step 3 of Scheme 3 the primary hydroxyl group is deprotected to lead to intermediate (III-d).
  • intermediate (III-c) is treated in the presence of an acid like para -toluenesulfonic acid in a alcoholic solvent like methanol at a temperature ranging from about -20 degrees Celsius to about room temperature to provide intermediate (III-d).
  • Cosolvents like chloroform may be used.
  • step 4 of Scheme 3 a hydroxymethylene group is introduced through a process similar to the one already described in Scheme 1 (step 1) and Scheme 2 (steps 4 and 5).
  • Other sources of formaldehyde like paraformaldehyde in a solvent like ethanol at a temperature ranging from about room temperature to about 70 degrees Celsius in the presence of an alkali metal alkoxide can also be used in this step.
  • Pg 4 is Bn
  • this step provides intermediate (III-e)
  • Pg 4 is Ac or Bz
  • this step provides intermediate (III-f).
  • intermediate (III-e) is treated with an acid like trifluoroacetic acid or an acidic resin in a solvent like dichloromethane at a temperature ranging from about -10 degrees Celsius to about room temperature to produce intermediate (III-g).
  • the remaining protecting groups (Pg 4 ) may then be removed using the appropriate chemistry for the particular protecting groups.
  • benzyl protecting groups may be removed by treating with formic acid in the presence of palladium (Pd black) in a protic solvent (e.g., ethanol/THF) at about room temperature to produce the final product (A).
  • a protic solvent e.g., ethanol/THF
  • step 7 of Scheme 3 intermediate (III-f) is treated with an acid like trifluoroacetic acid or an acidic resin in a solvent like dichloromethane at a temperature ranging from about -10 degrees Celsius to about room temperature to produce the final product (A).
  • step 1 of Scheme 4 intermediate (III-a) is treated with the appropriate arylsulfonyl chloride R 4 SO 2 Cl or arylsulfonic anhydride R 4 S(O) 2 OS(O) 2 R 4 (wherein R 4 is an optionally substituted aryl group, such as found in the arylsulfonyl chlorides 4-methyl-benzenesulfonyl chloride, 4-nitro-benzenesulfonyl chloride, 4-fluoro-benzenesulfonyl chloride, 2,6-dichloro-benzenesulfonyl chloride, 4-fluoro-2-methyl-benzenesulfonyl chloride, and 2,4,6-trichloro-benzenesulfonyl chloride, and in the arylsulfonic anhydride, p -toluenesulfonic anhydride) in presence of a base like pyridine, triethylamine, N , N -d
  • intermediate (IV-a) is submitted to a Kornblum-type oxidation (see, Kornblum, N., et al., Journal of The American Chemical Society, 81, 4113 (1959 )) to produce the corresponding aldehyde which may exist in equilibrium with the corresponding hydrate and/or hemiacetal form.
  • intermediate (IV-a) is treated in the presence of a base like pyridine, 2,6-lutidine, 2,4,6-collidine, N , N -diisopropylethylamine, 4-(dimethylamino)pyridine in a solvent like dimethyl sulfoxide at a temperature ranging from about room temperature to about 150 degrees Celsius.
  • the aldehyde intermediate produced is then submitted to the aldol/Cannizzaro conditions described for step 1 (Scheme 1) and step 5 (Scheme 2) to produce intermediate (IV-b).
  • step 3 of Scheme 4 intermediate (IV-b) is treated with an acid like trifluoroacetic acid or an acidic resin in a solvent like dichloromethane at a temperature ranging from about -10 degrees Celsius to about room temperature to produce the final product (A).
  • R 2 is (C 2 -C 4 )alkynyl
  • the process may be performed using Scheme 5, wherein R 6 is H or (C 1 -C 2 )alkyl.
  • step 1 of Scheme 5, which provides intermediate (V-i) the organometallic addition step is carried out in a similar way to the one described in Scheme1, step 6, using the organometallic reagent derived from (V-a), where Pg 5 is a suitable protective group for the hydroxyl group.
  • Pg 5 can be a tert -butyldimethylsilyl group (TBS) (see US2007/0054867 for preparation of for instance ⁇ 4-[(5-bromo-2-chloro-phenyl)-methyl]-phenoxy ⁇ - tert -butyl-dimethyl-silane).
  • protecting groups (Pg 5 ) and (Pg 1 ) can be removed to provide (V-k).
  • (Pg 5 ) is TBS and Pg 1 is Bn.
  • the protecting groups are removed by sequential treatment of (V-j) with 1) tetrabutylammonium fluoride in a solvent like tetrahydrofuran or 2-methyltetrahydrofuran at a temperature ranging from 0 degrees Celsius to about 40 degrees Celsius and 2) treatment with formic acid in the presence of palladium (Pd black) in a protic solvent (e.g., ethanol/THF) at about room temperature.
  • a protic solvent e.g., ethanol/THF
  • intermediate (V-k) is treated with N,N-bis-(trifluoromethanesulfonyl)-aniline in presence of a base like triethylamine or 4-dimethyaminopyridine in a solvent like dichloromethane or 1,2-dichloroethane at a temperature ranging from 0 degrees Celsius to about 40 degrees Celsius to produce intermediate (V-I).
  • a base like triethylamine or 4-dimethyaminopyridine in a solvent like dichloromethane or 1,2-dichloroethane at a temperature ranging from 0 degrees Celsius to about 40 degrees Celsius
  • step 5 of Scheme 5 intermediate (V-I) is subjected to a Sonogashira-type reaction (see, Sonogashira, K. Coupling Reactions Between sp2 and sp Carbon Centers. In Comprehensive Organic Synthesis (eds. Trost, B. M., Fleming, I.), 3, 521-549, (Pergamon, Oxford, 1991 )).
  • (V-I) is treated with the appropriate terminal alkyne HCCR 6 in presence of copper(I) iodide, a catalyst like bis-(triphenylphosphine)-palladium dichloride or tetrakis(triphenylphosphine)palladium(0) in presence of a base like triethylamine or N , N- diisopropylethylamine in a solvent like N , N -dimethylformamide at a temperature ranging from about room temperature to about 120 degrees Celsius to produce the desired product (A) and (B).
  • R 6 is H, it is more convenient to use trimethylsilylacetylene.
  • (V-k) represents different ways of accessing intermediate (V-k).
  • (V-k) can be treated with an alkylating agent of choice under classical conditions to selectively alkylate the phenol group to produce (A) (and (B) in schemes 1 and 5) where R 2 is (C 1 -C 4 )alkoxy.
  • the compounds of the present invention contain asymmetric or chiral centers, and, therefore, exist in different stereoisomeric forms. Unless specified otherwise, it is intended that all stereoisomeric forms of the compounds of the present invention as well as mixtures thereof, including racemic mixtures, form part of the present invention.
  • the present invention embraces all geometric and positional isomers. For example, if a compound of the present invention incorporates a double bond or a fused ring, both the cis - and trans- forms, as well as mixtures, are embraced within the scope of the invention.
  • Diastereomeric mixtures can be separated into their individual diastereoisomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization, distillation, sublimation.
  • Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereoisomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers.
  • an appropriate optically active compound e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride
  • some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention. Enantiomers can also be separated by use of a chiral HPLC (high pressure liquid chromatography) column.
  • HPLC high pressure liquid chromatography
  • tautomer or "tautomeric form” refers to structural isomers of different energies which are interconvertible via a low energy barrier.
  • proton tautomers also known as prototropic tautomers
  • proton tautomers include interconversions via migration of a proton, such as keto-enol and imine-enamine isomerizations.
  • a specific example of a proton tautomer is the imidazole moiety where the proton may migrate between the two ring nitrogens.
  • Valence tautomers include interconversions by reorganization of some of the bonding electrons. The equilibrium between closed and opened form of some intermediates (and/or mixtures of intermediates) is reminiscent of the process of mutarotation involving aldoses, known by those skilled in the art.
  • the present invention also embraces isotopically-labeled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, iodine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 31 P, 32 P, 35 S, 18 F, 123 I, 125 I and 36 Cl, respectively.
  • Certain isotopically-labeled compounds of the present invention are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e., 3 H) and carbon-14 (i.e., 14 C) isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances.
  • Positron emitting isotopes such as 15 O, 13 N, 11 C, and 18 F are useful for positron emission tomography (PET) studies to examine substrate occupancy.
  • Isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples herein below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • Compounds of the present invention are useful for treating diseases, conditions and/or disorders modulated by the inhibition of the sodium-glucose transporters (in particular SGLT2); therefore, another embodiment of the present invention is a pharmaceutical composition comprising a therapeutically effective amount of a compound of the present invention and a pharmaceutically acceptable excipient, diluent or carrier.
  • the compounds of the present invention (including the compositions and processes used therein) may also be used in the manufacture of a medicament for the therapeutic applications described herein.
  • a typical formulation is prepared by mixing a compound of the present invention and a carrier, diluent or excipient.
  • Suitable carriers, diluents and excipients are well known to those skilled in the art and include materials such as carbohydrates, waxes, water soluble and/or swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water, and the like.
  • the particular carrier, diluent or excipient used will depend upon the means and purpose for which the compound of the present invention is being applied. Solvents are generally selected based on solvents recognized by persons skilled in the art as safe (GRAS) to be administered to a mammal.
  • GRAS solvents recognized by persons skilled in the art as safe
  • safe solvents are non-toxic aqueous solvents such as water and other non-toxic solvents that are soluble or miscible in water.
  • Suitable aqueous solvents include water, ethanol, propylene glycol, polyethylene glycols (e.g., PEG400, PEG300), etc. and mixtures thereof.
  • the formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
  • buffers stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
  • the formulations may be prepared using conventional dissolution and mixing procedures.
  • the bulk drug substance i.e., compound of the present invention or stabilized form of the compound (e.g., complex with a cyclodextrin derivative or other known complexation agent)
  • a suitable solvent in the presence of one or more of the excipients described above.
  • the compound of the present invention is typically formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to give the patient an elegant and easily handleable product.
  • compositions also include solvates and hydrates of the compounds of Formula (I).
  • solvate refers to a molecular complex of a compound represented by Formula (I) (including pharmaceutically acceptable salts thereof) with one or more solvent molecules.
  • solvent molecules are those commonly used in the pharmaceutical art, which are known to be innocuous to the recipient, e.g., water, ethanol, ethylene glycol, and the like
  • hydrate refers to the complex where the solvent molecule is water.
  • the solvates and/or hydrates preferably exist in crystalline form.
  • solvents may be used as intermediate solvates in the preparation of more desirable solvates, such as methanol, methyl t-butyl ether, ethyl acetate, methyl acetate, (S)-propylene glycol, (R)-propylene glycol, 1,4-butyne-diol, and the like.
  • the crystalline forms may also exist as complexes with other innocuous small molecules, such as L-phenylalanine, L-proline, L-pyroglutamic acid and the like, as co-crystals or solvates or hydrates of the co-crystalline material.
  • the solvates, hydrates and co-crystalline compounds may be prepared using procedures described in PCT Publication No. WO 08/002824 , incorporated herein by reference, or other procedures well-known to those of skill in the art.
  • the pharmaceutical composition (or formulation) for application may be packaged in a variety of ways depending upon the method used for administering the drug.
  • an article for distribution includes a container having deposited therein the pharmaceutical formulation in an appropriate form.
  • Suitable containers are well-known to those skilled in the art and include materials such as bottles (plastic and glass), sachets, ampoules, plastic bags, metal cylinders, and the like.
  • the container may also include a tamper-proof assemblage to prevent indiscreet access to the contents of the package.
  • the container has deposited thereon a label that describes the contents of the container. The label may also include appropriate warnings.
  • the present invention further provides a use of the compounds of the present invention in a method of treating diseases, conditions and/or disorders modulated by the inhibition of sodium-glucose transporters in an animal that includes administering to an animal in need of such treatment a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition comprising an effective amount of a compound of the present invention and a pharmaceutically acceptable excipient, diluent, or carrier.
  • the method is particularly useful for treating diseases, conditions and/or disorders that benefit from the inhibition of SGLT2.
  • One aspect of the present invention is the treatment of obesity, and obesity-related disorders (e.g., overweight, weight gain, or weight maintenance).
  • obesity-related disorders e.g., overweight, weight gain, or weight maintenance.
  • BMI body mass index
  • Overweight is typically defined as a BMI of 25-29.9 kg/m 2
  • obesity is typically defined as a BMI of 30 kg/m 2 .
  • Another aspect of the present invention is for the treatment or delaying the progression or onset of diabetes or diabetes-related disorders including Type 1 (insulin-dependent diabetes mellitus, also referred to as “IDDM”) and Type 2 (noninsulin-dependent diabetes mellitus, also referred to as “NIDDM”) diabetes, impaired glucose tolerance, delayed wound healing, hyperinsulinemia, elevated blood levels of fatty acids, hyperlipidemia, hypertriglyceridemia, Syndrome X, increased high density lipoprotein levels, insulin resistance, hyperglycemia, and diabetic complications (such as atherosclerosis, coronary heart disease, stroke, peripheral vascular disease, nephropathy, hypertension, neuropathy, and retinopathy).
  • IDDM insulin-dependent diabetes mellitus
  • NIDDM noninsulin-dependent diabetes mellitus
  • Metabolic syndrome includes diseases, conditions or disorders such as dyslipidemia, hypertension, insulin resistance, diabetes (e.g., Type 2 diabetes), coronary artery disease and heart failure.
  • diabetes e.g., Type 2 diabetes
  • Metabolic Syndrome see, e.g., Zimmet, P.Z., et al., "The Metabolic Syndrome: Perhaps an Etiologic Mystery but Far From a Myth - Where Does the International Diabetes Federation Stand?,” Diabetes & Endocrinology, 7(2), (2005 ); and Alberti, K.G., et al., "The Metabolic Syndrome - A New Worldwide Definition,” Lancet, 366, 1059-62 (2005 ).
  • administration of the compounds of the present invention provides a statistically significant (p ⁇ 0.05) reduction in at least one cardiovascular disease risk factor, such as lowering of plasma leptin, C-reactive protein (CRP) and/or cholesterol, as compared to a vehicle control containing no drug.
  • cardiovascular disease risk factor such as lowering of plasma leptin, C-reactive protein (CRP) and/or cholesterol
  • the administration of compounds of the present invention may also provide a statistically significant (p ⁇ 0.05) reduction in glucose serum levels.
  • a dosage in the range of from about 0.001 mg to about 10 mg per kilogram body weight is typically sufficient, preferably from about 0.01 mg/kg to about 5.0 mg/kg, more preferably from about 0.01 mg/kg to about 1 mg/kg.
  • some variability in the general dosage range may be required depending upon the age and weight of the subject being treated, the intended route of administration, the particular compound being administered and the like.
  • the determination of dosage ranges and optimal dosages for a particular patient is well within the ability of one of ordinary skill in the art having the benefit of the instant disclosure.
  • the compounds of the present invention can be used in sustained release, controlled release, and delayed release formulations, which forms are also well known to one of ordinary skill in the art.
  • the compounds of this invention may also be used in conjunction with other pharmaceutical agents for the treatment of the diseases, conditions and/or disorders described herein. Therefore, methods of treatment that include administering compounds of the present invention in combination with other pharmaceutical agents are also provided.
  • Suitable pharmaceutical agents that may be used in combination with the compounds of the present invention include anti-obesity agents (including appetite suppressants), anti-diabetic agents, anti-hyperglycemic agents, lipid lowering agents, anti-inflammatory agents and anti-hypertensive agents.
  • Suitable anti-obesity agents include cannabinoid-1 (CB-1) antagonists (such as rimonabant), 11 ⁇ -hydroxy steroid dehydrogenase-1 (11 ⁇ -HSD type 1) inhibitors, stearoyl-CoA desaturase-1 (SCD-1) inhibitor, MCR-4 agonists, cholecystokinin-A (CCK-A) agonists, monoamine reuptake inhibitors (such as sibutramine), sympathomimetic agents, ⁇ 3 adrenergic agonists, dopamine agonists (such as bromocriptine), melanocyte-stimulating hormone analogs, 5HT2c agonists, melanin concentrating hormone antagonists, leptin (the OB protein), leptin analogs, leptin agonists, galanin antagonists, lipase inhibitors (such as tetrahydrolipstatin, i.e.
  • CBD-1 cannabinoid-1
  • anorectic agents such as a bombesin agonist
  • neuropeptide-Y antagonists e.g., NPY Y5 antagonists
  • PYY 3-36 including analogs thereof
  • thyromimetic agents dehydroepiandrosterone or an analog thereof
  • glucocorticoid agonists or antagonists orexin antagonists
  • glucagon-like peptide-1 agonists ciliary neurotrophic factors (such as AxokineTM available from Regeneron Pharmaceuticals, Inc., Tarrytown, NY and Procter & Gamble Company, Cincinnati, OH)
  • human agouti-related protein (AGRP) inhibitors ghrelin antagonists, histamine 3 antagonists or inverse agonists
  • neuromedin U agonists e.g., MTP/ApoB inhibitors (e.g., gut-selective MTP inhibitors, such as dirlotapide), opioid antagonist, orexin antagonist, and the like.
  • Preferred anti-obesity agents for use in the combination aspects of the present invention include CB-1 antagonists (e.g., rimonabant, taranabant, surinabant, otenabant, SLV319 (CAS No. 464213-10-3) and AVE1625 (CAS No. 358970-97-5)), gut-selective MTP inhibitors (e.g., dirlotapide, mitratapide and implitapide, R56918 (CAS No. 403987) and CAS No.
  • CB-1 antagonists e.g., rimonabant, taranabant, surinabant, otenabant, SLV319 (CAS No. 464213-10-3) and AVE1625 (CAS No. 358970-97-5)
  • gut-selective MTP inhibitors e.g., dirlotapide, mitratapide and implitapide, R56918 (CAS No. 403987) and
  • CCKa agonists e.g., N-benzyl-2-[4-(1H-indol-3-ylmethyl)-5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-tetraaza-benzo[e]azulen-6-yl]-N-isopropyl-acetamide described in PCT Publication No. WO 2005/116034 or US Publication No.
  • PYY 3-36 includes analogs, such as peglated PYY 3-36 e.g., those described in US Publication 2006/0178501 ), opioid antagonists (e.g., naltrexone), oleoyl-estrone (CAS No.
  • compounds of the present invention and combination therapies are administered in conjunction with exercise and a sensible diet.
  • Suitable anti-diabetic agents include an acetyl-CoA carboxylase-2 (ACC-2) inhibitor, a phosphodiesterase (PDE)-10 inhibitor, a diacylglycerol acyltransferase (DGAT) 1 or 2 inhibitor, a sulfonylurea (e.g., acetohexamide, chlorpropamide, diabinese, glibenclamide, glipizide, glyburide, glimepiride, gliclazide, glipentide, gliquidone, glisolamide, tolazamide, and tolbutamide), a meglitinide, an ⁇ -amylase inhibitor (e.g., tendamistat, trestatin and AL-3688), an ⁇ -glucoside hydrolase inhibitor (e.g., acarbose), an ⁇ -glucosidase inhibitor (e.g., adiposine, camiglibose,
  • Suitable anti-inflammatory agents include genital tract/urinary tract infection preventatives and treatments.
  • exemplary agents include cranberries (i.e. Vaccinium macrocarpon ) and cranberry derivatives such as cranberry juice, cranberry extracts or flavonols of cranberries.
  • Cranberry extracts may include one or more flavonols (i.e.
  • anthocyanins and proanthocyanidins or a purified cranberry flavonol compound, including myricetin-3- ⁇ -xylopyranoside, quercetin-3- ⁇ -glucoside, quercetin-3- ⁇ -arabinopyranoside, 3'-methoxyquercetin-3- ⁇ -xylopyranoside, quercetin-3-O-(6"-p-coumaroyl)- ⁇ -galactoside, quercetin-3-O-(6"-benzoyl)- ⁇ -galactoside, and/or quercetin-3- ⁇ -arabinofuranoside.
  • starting materials are generally available from commercial sources such as Aldrich Chemicals Co. (Milwaukee, WI), Lancaster Synthesis, Inc. (Windham, NH), Acros Organics (Fairlawn, NJ), Maybridge Chemical Company, Ltd. (Cornwall, England), Tyger Scientific (Princeton, NJ), AstraZeneca Pharmaceuticals (London, England), and Accela ChemBio (San Diego, CA).
  • NMR spectra were recorded on a Varian UnityTM 400 (available from Varian Inc., Palo Alto, CA) at room temperature at 400 MHz for proton. Chemical shifts are expressed in parts per million (delta) relative to residual solvent as an internal reference.
  • the peak shapes are denoted as follows: s, singlet; d, doublet; dd, doublet of doublet; t, triplet; q, quartet; m, multiplet; bs or br.s., broad singlet; 2s, two singlets; br.d., broad doublet.
  • Electrospray ionization mass spectra were obtained on a WatersTM ZMD instrument (carrier gas: nitrogen; solvent A: water /0.01% formic acid, solvent B: acetonitrile/0.005% formic acid; available from Waters Corp., Milford, MA).
  • High resolution mass spectra HRMS were obtained on an AgilentTM Model 6210 time of flight. Where the intensity of single chlorine or single bromine-containing ions are described, the expected intensity ratio was observed (approximately 3:1 for 35 Cl/ 37 Cl-containing ions and 1:1 for 79 Br/ 81 Br-containing ions) and the intensity of only the lower mass ion is given. In some cases only representative 1 H NMR peaks are given.
  • HPLC high pressure liquid chromatography
  • ShimadzuTM 10A LC-UV or a AgilentTM 1100 preparatory HPLC were performed using a ShimadzuTM 10A LC-UV or a AgilentTM 1100 preparatory HPLC. Except where otherwise noted, all reactions were run under an inert atmosphere of nitrogen gas using anhydrous solvents. Also, except where otherwise noted, all reactions were run at room temperature ( ⁇ 23°C).
  • R f is defined as the ratio of the distance traveled by the compound divided by the distance traveled by the eluent.
  • R t retention time).
  • any of the following starting materials can be prepared using the procedures described in Schemes 7 or 8 of US Publication No. 2008/0132563 , or alternatively, Schemes 2, 3 or 8 of US Publication No. 2007/0259821 . More specifically, the following starting materials used in the following Examples can be prepared using the procedures described in the corresponding references or purchased from the corresponding vendor.
  • 4-Bromo-2-(4-methoxy-benzyl)-1-methyl-benzene can be prepared by the procedures described in Example 8 of PCT Publication No. WO 01/027128 .
  • 4-Bromo-2-(4-ethoxy-benzyl)-1-methyl-benzene can be prepared by the procedures described in Preparation Example 17 of US2008/0132563 .
  • 4-Bromo-1-chloro-2-(4-methoxy-benzyl)-benzene can be prepared by the procedures described in Preparation Example 19 of US2008/0132563 or Example V of US2007/0259821 .
  • 4-Bromo-1-chloro-2-(4-ethoxy-benzyl)-benzene may be purchased from Shanghai Haoyuan Chemexpress Co., Ltd., Shanghai, People's Republic of China.
  • Oxalyl chloride (11.0 mL, 126 mmol) was added dropwise to a well stirred suspension of 5-bromo-2-fluoro-benzoic acid (25.0 g, 114 mmol) in dichloromethane (150 mL) and N , N -dimethylformamide (1.5 mL) at 0°C. The resulting mixture was allowed to gradually warm to room temperature. After 18 hours, the solid had gone into solution. The resulting light orange solution was concentrated under reduced pressure and was chased two times with diethyl ether to afford 5-bromo-2-fluoro-benzoyl chloride (27.0 g, quantitative yield) as a pale orange oil.
  • the organic phase was decanted and the aqueous residue was concentrated under reduced pressure to remove any remaining organic solvent.
  • the aqueous phase was extracted with ethyl acetate (200 mL ⁇ 2) and the extracts were combined with the decanted tetrahydrofuran solution. This solution was washed with brine (25 mL) and was dried (sodium sulfate), filtered and concentrated under reduced pressure, giving crude (5-bromo-2-fluorophenyl)-(4-chlorophenyl)-methanol (15.2 g, 96% yield) as a yellow solid.
  • reaction mixture was diluted with brine (2 L) and extracted with ethyl acetate (2 L).
  • the organic phases from the seven reactions were combined, evaporated and the residue purified on silica gel (eluting with 4 to 1 up to 1 to 1 hexanes in ethyl acetate) to give intermediate compound ( I-1b ) as a mixture of anomers (980 g, 53% yield over the two steps).
  • R f 0.57 and 0.60 in 1:1 hexanes/ethyl acetate.
  • This compound was prepared starting from [((3S,4S,5R)-6-allyloxy-3,4,5-tris-benzyloxy-2-hydroxymethyl-tetrahydro-pyran-2-yl)-methanol ( I-1b ) using a similar procedure as the one described for the synthesis of (2R,3S,4S)-2,3,4-tris-benzyloxy-5-hydroxy-6-(4-methoxy-benzyloxy)-5-(4-methoxy-benzyloxymethyl)-hexanoic acid methoxy-methyl-amide ( I-1g ) and/or (3R,4S,5S)-3,4,5-tris-benzyloxy-6,6-bis-(4-methoxy-benzyloxymethyl)-2-(methoxy-methyl-amino)-tetrahydro-pyran-2-ol ( I-1f ) except that the alkylating agent used in the experimental part describing the conversion from ( I-1 b ) to ( I-1c )
  • n -Butyl lithium (0.97 mL, 2.5 M/hexanes, 3.15 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution (placed in a pre dried Biotage TM microwave vial 10-20 mL sealed with its cap and placed under a positive stream of nitrogen gas) of 4-bromo-2-(4-methoxy-benzyl)-1-methyl-benzene (690 mg, 3 equivalents) in anhydrous tetrahydrofuran (2.7 mL) at -78°C and the resulting solution was stirred at this temperature for an additional hour.
  • n -Butyl lithium (0.312 mL, 2.5 M/hexanes, 3.05 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution (placed in a pre dried Biotage TM microwave vial 10-20 mL sealed with its cap and placed under a positive stream of nitrogen gas) of 4-bromo-2-(4-ethoxy-benzyl)-1-methyl-benzene (238 mg, 3.05 equivalents) in anhydrous tetrahydrofuran (0.9 mL) at -78°C and the resulting solution was stirred at this temperature for an additional hour.
  • n-Butyl lithium (0.97 mL, 2.5 M/hexanes, 3.15 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution (placed in a pre dried Biotage TM microwave vial 10-20 mL sealed with its cap and placed under a positive stream of nitrogen gas) of 4-bromo-1-chloro-2-(4-methoxy-benzyl)-benzene (725 mg, 2.95 equivalents) in anhydrous tetrahydrofuran (2.7 mL) at -78°C and the resulting solution was stirred at this temperature for an additional hour.
  • n -Butyl lithium (1.0 mL, 2.5 M/hexanes, 3.25 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution (placed in a pre dried Biotage TM microwave vial 10-20 mL sealed with its cap and placed under a positive stream of nitrogen gas) of 4-bromo-1-chloro-2-(4-ethoxy-benzyl)-benzene (815 mg, 3.25 equivalents) in anhydrous tetrahydrofuran (2.9 mL) at -78°C and the resulting solution was stirred at this temperature for an additional hour.
  • n-Butyl lithium (462 microL, 2.5 M/hexanes, 3.0 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution (placed in a pre dried Biotage TM microwave vial 10-20 mL sealed with its cap and placed under a positive stream of nitrogen gas) of 4-Bromo-1-fluoro-2-(4-methoxy-benzyl)-benzene (341 mg, 3 equivalents) in anhydrous tetrahydrofuran (1.4 mL) at -78 °C under nitrogen. The resulting solution was stirred at this temperature for 1 hour.
  • the reaction was diluted with diethyl ether and quenched by the dropwise addition of aqueous 1 N hydrochloric acid solution.
  • the resulting biphasic mixture was stirred at room temperature for 15 minutes.
  • the organic phase was separated, washed with brine, dried over magnesium sulfate, filtered, and concentrated under reduced pressure.
  • the crude residue was purified by flash chromatography over silica gel (eluting with a gradient of 10 to 40% ethyl acetate in heptane) to afford the product as a mixture of isomers (199 mg, 55 % yield).
  • n-Butyl lithium (508 microL, 2.5 M/hexanes, 3.0 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution of 4-Bromo-2-(4-ethoxy-benzyl)-1-fluorobenzene (392.0 mg, 1.27 mmol) in anhydrous tetrahydrofuran (1.5 mL) at -78°C under nitrogen. The resulting solution was stirred at this temperature for 1 hour.
  • the reaction was diluted with diethyl ether and quenched by the dropwise addition of aqueous 1 N hydrochloric acid solution.
  • the resulting biphasic mixture was stirred at room temperature for 15 minutes.
  • the organic phase was separated, washed with brine, dried over magnesium sulfate, filtered, and concentrated under reduced pressure.
  • the crude residue was purified by flash chromatography over silica gel (eluting with a gradient of 10 to 40% ethyl acetate in heptane) to afford the product as a mixture of isomers (180 mg, 44% yield).
  • n-Butyl lithium (1.0 mL, 2.5 M/hexanes, 3.0 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution of 3-[4-(5-bromo-2-fluoro-benzyl)-phenoxy]-tetrahydro-furan (878 mg, 2.50 mmol) in anhydrous tetrahydrofuran (3.0 mL) at -78°C and the resulting solution was stirred at this temperature for 1 hour.
  • n-Butyl lithium (1.0 mL, 2.5 M/hexanes, 3.1 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution (placed in a pre dried Biotage TM microwave vial 10-20 mL sealed with its cap and placed under a positive stream of nitrogen gas) of 4-Bromo-2-(4-chloro-benzyl)-1-fluoro-benzene (702 mg, 2.9 equivalents) in anhydrous tetrahydrofuran (3.0 mL) at -78°C and the resulting solution was stirred at this temperature for 25 minutes.
  • n-Butyl lithium (1.12 mL, 2.5 M/hexanes, 3.0 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution of 3-[4-(5-Bromo-2-fluoro-benzyl)-phenoxy]-oxetane (942.0 mg, 2.79 mmol) in anhydrous tetrahydrofuran (3.0 mL) at -78°C and the resulting solution was stirred at this temperature for 1 hour.
  • the reaction was diluted with diethyl ether and quenched by the dropwise addition of aqueous 1 N hydrochloric acid solution.
  • the resulting biphasic mixture was stirred at room temperature for 15 minutes.
  • the organic phase was separated, washed with brine, dried over magnesium sulfate, filtered, and concentrated under reduced pressure.
  • the crude residue was purified by flash chromatography over silica gel (eluting with a gradient of 10 to 40% ethyl acetate in heptane) to afford the product as a mixture of isomers (535 mg, 59 % yield).
  • n-Butyl lithium (0.97 mL, 2.5 M/hexanes, 3.15 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution (placed in a pre dried Biotage TM microwave vial 10-20 mL sealed with its cap and placed under a positive stream of nitrogen gas) of 3-(4-(5-bromo-2-chlorobenzyl)phenoxy)oxetane (824 mg, 2.95 equivalents) in anhydrous tetrahydrofuran (2.7 mL) at -78°C and the resulting solution was stirred at this temperature for an additional hour.
  • the palladium was filtered and the crude mixture obtained after evaporation of solvent was purified by chromatography over silica gel eluting with a gradient of 85 to 100% ethyl acetate in heptane.
  • the mixture of products obtained was purified by HPLC preparative.
  • HPLC preparative method reverse phase C18 phenomenex column Luna 5 micrometer 150 x 21.20 mm, 20 mL/minute, gradient of acetonitrile/0.1 % formic acid:water/0.1 % formic acid; 20 to 60% of acetonitrile/0.1 % formic acid over 20 minutes. UV detection: 254 nm.
  • HPLC preparative method reverse phase C18 phenomenex column Luna 5 micrometer 150 x 21.20 mm, 20 mL/minute, gradient of acetonitrile/0.1 % formic acid:water/0.1 % formic acid; 20 to 60% of acetonitrile/0.1 % formic acid over 20 minutes. UV detection: 254 nm.
  • HPLC preparative method reverse phase C18 phenomenex column Luna 5 micrometer 150 x 21.20 mm, 20 mL/minutes, gradient of acetonitrile/0.1 % formic acid:water/0,1 % formic acid; 20 to 60% of acetonitrile/0.1 %formic acid over 20 minutes. UV detection: 254 nm.
  • HPLC preparative reverse phase C18 Gemini column 5 micrometer 30 x 100 mm, 40 mL/minute, gradient of acetonitrile/0.1 % formic acid : water/0.1 % formic acid; 25 to 50% of acetonitrile/0.1 % formic acid over 18 minutes; UV detection: 220 nm.
  • HPLC preparative method reverse phase C18 phenomenex column Luna 5 micrometer 150 x 21.20 mm, 20 mL/minute, gradient of acetonitrile/0.1 % formic acid:water/0.1 % formic acid; 20 to 80% of acetonitrile/0.1 % formic acid over 20 minutes). UV detection: 254 nm.
  • n-Butyl lithium (1.04 mL, 2.6 mmol, 2.5 M in hexane) was added to a solution of isopropyl magnesium bromide (1.27 mL, 1.27 mmol, 1 M in tetrahydrofuran) at 0 °C. After being stirred for 30 minutes, the resulting mixture was cooled to -78°C and a solution of 4-bromo-2-(4-ethoxy-benzyl)-benzonitrile (380 mg, 1.20 mmol) in anhydrous tetrahydrofuran (1 mL) was added.
  • Example 8 illustrates the preparation of a crystalline derivative of the compound of Example 3B in order to confirm the structure and stereochemistry of Example 3B.
  • a trial structure was obtained by direct methods. This trial structure refined routinely. Hydrogen positions were calculated wherever possible. The methyl hydrogens were located by difference Fourier techniques and then idealized. The hydrogen parameters were added to the structure factor calculations but were not refined. The shifts calculated in the final cycles of least squares refinement were all less than 0.1 of the corresponding standard deviations. The final R-index was 3.71 %. A final difference Fourier revealed no missing or misplaced electron density. The refined structure was plotted using the SHELXTL plotting package ( Figure 1 ). The absolute configuration was determined by the method of Flack. See, Flack, H.D., Acta Crystallogr ., A39 , 876, (1983).
  • Example 9 illustrates the preparation of a crystalline derivative of the compound of Example 4A in order to confirm the structure and stereochemistry of Example 4A.
  • a trial structure was obtained by direct methods. This trial structure refined routinely. Hydrogen positions were calculated wherever possible. The methyl hydrogens were located by difference Fourier techniques and then idealized. The hydrogen parameters were added to the structure factor calculations but were not refined. The shifts calculated in the final cycles of least squares refinement were all less than 0.1 of the corresponding standard deviations. The final R-index was 4.36%. A final difference Fourier revealed no missing or misplaced electron density.
  • the refined structure was plotted using the SHELXTL plotting package ( Figure 2 ).
  • the absolute configuration was determined by the method of Flack See, Flack, H.D., Acta Crystallogr ., A39 , 876, (1983).
  • HPLC preparative method reverse phase C18 Gemini column, 5 micrometer 30x100mm, 40mL/minute flow rate, gradient of acetonitrile/0.1 % formic acid : water/0.1 % formic acid; 25 to 50% acetonitrile/0.1 % formic acid over 18 minutes; UV detection: 220 nm.
  • HPLC analytical method reverse phase C18 Gemini column, 5 ⁇ m 4.6x150mm, 1 mL/minute flow rate, gradient of acetonitrilel0.1 % trifluoroacetic acid : water/0.1 % trifluoroacetic acid; 5 to 100% acetonitrile/0.1 % trifluoroacetic acid over 12 minutes; UV detection: 220 nm.
  • HPLC preparative method reverse phase C18 Gemini column, 5 micrometer 30x100mm, 40mL/minute flow rate, gradient of acetonitrile/0.1 % formic acid : water/0.1 % formic acid; 25 to 50% acetonitrile/0.1 % formic acid over 18 minutes; UV detection: 220 nm.
  • HPLC analytical method reverse phase C18 Gemini column, 5 micrometer 4.6x150mm, 1 mL/minute flow rate, gradient of acetonitrile/0.1 % trifluoroacetic acid : water/0.1 % trifluoroacetic acid; 5 to 100% acetonitrile/0.1 % trifluoroacetic acid over 12 minutes; UV detection: 220 nm.
  • HPLC preparative conditions reverse phase C18 Gemini column 5 micrometer 30 x 100 mm, flow rate 40 mL/minute, gradient of acetonitrile/0.1 % formic acid : water/0.1 % formic acid; 25 to 50% of acetonitrile/0.1 % formic acid over 18 minutes), UV detection: 220 nm.
  • HPLC analytical method reverse phase C18 Gemini column, 5 ⁇ m 4.6x150mm, 1 mL/minute flow rate, gradient of acetonitrile/0.1 % trifluoroacetic acid : water/0.1 % trifluoroacetic acid; 5 to 100% acetonitrile/0.1 % trifluoroacetic acid over 12 minutes; UV detection: 220 nm.
  • HPLC preparative method reverse phase C18 Gemini column, 5 micrometer 30x100mm, 40mL/minute flow rate, gradient of acetonitrile/0.1 % formic acid : water/0.1 % formic acid; 25 to 50% acetonitrile/0.1 % formic acid over 18 minutes; UV detection: 220 nm.
  • HPLC preparative method reverse phase C18 Xbridge column 5 micrometer 100 x 30 mm, flow rate 40 mL/minute, gradient of acetonitrile/0.1 % formic acid : water/0.1 %formic acid; 30 to 55% of acetonitrile/0.1 % formic acid over 11 minutes; UV detection: 220 nm.
  • L-proline dissolved in water (approximately 480 mg/mL) was added to the example 4A compound (approximately 80 moles L-proline per mole (example 4A compound)).
  • Volume was doubled with ethanol and solution was capped and stirred for approximately 12 hours. Volume was reduced by half by evaporation on the bench. Volume was doubled using ethanol and the volume of solution was again reduced by half using evaporation. Solid was recovered using centrifugal filtration.
  • L-proline dissolved in water (approximately 480 mg/mL) was added to the example 4A compound (approximately 59 moles L-proline per mole example 4A compound).
  • Volume was doubled with methanol and solution was clear. Volume was increased by 25% using acetone. Solution was capped and stirred for approximately 12 hours. Volume was reduced by approximately 60% through evaporation on the bench. Volume was doubled using methanol and remaining solvent was evaporated leaving solid white precipitate.
  • a solution of ethanol saturated with L-proline was added to the example 4A compound (approximately 2.2 moles L-proline per mole example 4A compound) in a glass vial. Clear solution was capped and stirred for approximately 72 hours. Volume was reduced by half by evaporation at room temperature. Precipitate was seen and vial was capped and stirred for approximately 12 hours. White solid was collected using centrifugal filtration.
  • Example 4A compound was added to 3.2 mL of the L-pyroglutamic acid solution. After approximately 1 minute, precipitation was seen. The solution was too thick to stir therefore 2 mL of a 1:1 ethanol/water solution was added. The solution was stirred overnight. The solid was collected using vacuum filtration on a 0.45 microm nylon filter membrane. The solid was dried in a 50 °C vacuum oven for approximately 2 hours. Approximately 960 mg of the cocrystal complex (20) was recovered. The stoichiometric ratio of Example 4A compound to L-pyroglutamic acid was determined using quantitative NMR to be 1:1.63. Excess L-pyroglutamic acid was removed by suspending the material in ethanol yielding 1:1 co-crystal (20).
  • Powder X-ray Diffraction Analysis The powder X-ray diffraction patterns of the cocrystal of example 4A compound with L-proline and the cocrystal of example 4A compound with L-pyroglutamic acid were carried out on a Bruker D5000 diffractometer using copper radiation (wavelength: 1.54056 ⁇ ). The tube voltage and amperage were set to 40 kV and 40mA, respectively. The divergence and scattering slits were set at 1 mm, and the receiving slit was set at 0.6 mm. Diffracted radiation was detected by a Kevex PSI detector.
  • the sample is typically placed into a holder which has a cavity.
  • the sample powder is pressed by a glass slide or equivalent to ensure a random surface and proper sample height.
  • the sample holder is then placed into the instrument.
  • the incident X-ray beam is directed at the sample, initially at a small angle relative to the plane of the holder, and then moved through an arc that continuously increases the angle between the incident beam and the plane of the holder.
  • Measurement differences associated with such X-ray powder analyses result from a variety of factors including: (a) errors in sample preparation (e.g., sample height), (b) instrument errors (e.g.
  • shifts can be identified from the X-ray Diffractogram and can be eliminated by compensating for the shift (applying a systematic correction factor to all peak position values) or recalibrating the instrument.
  • a systematic correction factor to bring the peak positions into agreement.
  • this correction factor will bring the measured peak positions from the Bruker into agreement with the expected peak positions and may be in the range of 0 to 0.2 ° 2 ⁇ .
  • the powder X-ray diffraction values are generally accurate to within ⁇ 0.2 2-theta degrees, due to slight variations of instrument and test conditions.
  • Cocrystal of example 4A compound and L-proline from Example 18 characterized by the following powder x-ray diffraction pattern, provided in Figure 3 , expressed in terms of the degree 2 ⁇ and relative intensities with a relative intensity of ⁇ 2.7% measured on a Bruker D5000 diffractometer with CuK ⁇ radiation: Angle (Degree 2 ⁇ ) Relative Intensity* ( ⁇ 2.7%) 4.6 5.1 5.5 12.8 7.6 40.2 8.5 11.9 10.3 9.1 11.0 4.5 12.1 22.3 12.6 13.5 14.4 13.1 14.8 16.1 15.3 2.7 15.9 10.4 16.5 3.0 16.8 8.2 17.0 16.6 17.4 33.9 18.1 2.9 18.4 10.3 18.9 16.8 19.5 12.2 20.3 100.0 21.0 6.5 22.0 5.5 22.2 7.1 22.6 11.5 22.9 29.3 23.5 4.5 24.3 13.8 24.8 14.2 25.4 14.7 2.5.7 23.2 26.0 6.9 26.8 5.9 27.0 5.8 27.5 21.2 28.8 15.5 29.4 6.5 29.
  • Characteristic 2 ⁇ peaks or combinations of cocrystal of example 4A compound and L-proline Angle (Degree 2 ⁇ ) 7.6 12.1 20.3 28.8
  • Cocrystal of example 4A compound and L-pyroglutamic acid from Example 20 characterized by the following powder x-ray diffraction pattern, provided in Figure 4 , expressed in terms of the degree 2 ⁇ and relative intensities with a relative intensity of ⁇ 2.7% measured on a Bruker D5000 diffractometer with CuK ⁇ radiation: Angle (Degree 2 ⁇ ) Relative Intensity* ( ⁇ 4.3%) 6.4 31.0 7.6 5.9 11.8 4.3 12.3 8.8 12.7 11.0 13.5 10.4 14.2 31.3 14.4 24.0 15.2 13.9 16.7 66.8 17.4 57.1 18.3 10.5 18.7 53.3 19.1 24.2 19.3 32.0 19.8 9.2 20.3 75.6 21.1 100.0 22.5 9.2 23.6 11.7 24.3 18.7 24.7 22.2 25.0 14.2 26.2 53.4 27.2 4.6 27.9 10.0 28.3 26.3 29.0 14.5 29.5 31.3 30.7 16.2 31.5 5.4 32.0 23.2 33.0 9.9 34.2 19.9 35.2 5.4 35.9 1
  • Thermograms were obtained on a TA Instruments Q1000 Differential Scanning Calorimeter (DSC). 1-2 mg of sample was placed in aluminum sample pans and then covered with a pierced lid. The energy was measured against an empty pan as the temperature increased from 25°C to 200-300°C at 10°C per minute. The onset temperature of the melting endotherm was reported as the melting temperature. The onset temperature of the melting endotherm is dependent on the rate of heating, the purity of the sample, size of crystal and sample, among other factors. Typically, the DSC results are accurate to within about ⁇ 2°C, preferably to within ⁇ 1.5°C.
  • Example 18 cocrystal of Example 4A compound and L-proline DSC results are shown in Figure 5 .
  • Example 20 cocrystal of Example 4A compound and L-pyroglutamic acid DSC results are shown in Figure 6 .
  • the relevant hydrogen atoms bonded to 05 were located from Fourier techniques, but was deleted and placed in idealized location (HFIX 83).
  • the relevant hydrogen atom bonded to 04 could not be found with Fourier techniques and was placed in an idealized location (HFIX 83).
  • the hydrogen atoms on the water molecule could not be located and were left out of the solution.
  • the hydrogen parameters were added to the structure factor calculations but were not refined.
  • the shifts calculated in the final cycles of least squares refinement were all less than 0.1 of the corresponding standard deviations.
  • the final R-index was 5.15%.
  • a final difference Fourier revealed no missing or misplaced electron density.
  • the refined structure was plotted using the SHELXTL plotting package ( Figure 7 ).
  • the absolute configuration was determined by the method of Flack 4 . Coordinates, anisotropic temperature factors, distances and angles are available as supplementary material (Tables 24-2 to 24-5).
  • Anisotropic displacement parameters ( ⁇ 2 x 10 3 ) for Example 24.
  • the anisotropic displacement factor exponent takes the form: -2 ⁇ 2 [ h 2 a* 2 U 11 + ... + 2 h k a* b* U 12 ] U 11 U 22 U 33 U 23 U 13 U 12 CI(01) 77(1) 69(1) 69(1) -11(1) 1(1) -27(1) N(1) 56(2) 40(2) 60(2) 0(2) 12(2) 0(2) O(1) 70(2) 70(3) 62(2) 2(2) 23(2) -3(2) C(1) 80(3) 76(4) 77(3) -2(3) 13(3) 8(3) O(2) 47(1) 52(2) 49(1) -1(1) 3(1) 12(1) C(2) 63(3) 84(4) 52(2) 1(2) 13(2) 7(3) O(3) 46(1) 43(2) 45(1) 2(1) 3(1) -1(1) C(3) 48(2) 59(3) 57(2) 5(2) 12(2) 8(2) O(4) 111(3) 83(3) 62(2) 8(2) -10(2) 23(3) C(4) 59(3) 72(4) 53(2) -10(2) 10(2) -12(2) C(5) 54(2) 66
  • a trial structure was obtained by direct methods. This trial structure refined routinely, except for a low residual peak that was refined as 0.1 stoichiometric water.
  • the stoichiometry of water was found by first deleting a hydroxyl group on the molecule, refining and measuring the resulting q peak; then comparing this peak to the residual peak from the water molecule. Using this method, a ratio of 1 to 0.1 (molecule to water) was estimated. Additionally, removing the water molecule from the solution and searching for void spaces in the crystal with Material Studio, Platon, and Mercury revealed a plausible volume of 33 cubic angstroms for a water molecule (water typically has about 40 cubic angstroms of space).
  • the hydrogen atoms on nitrogen and oxygen were located by difference Fourier techniques and allowed to refine freely with no restraints.
  • a few of the protons bonded to heteroatoms (H97a, H97b, H97c, and H97c) exhibited somewhat short bond lengths ( ⁇ 0.8 angstroms found vs. ⁇ 0.96 expected), yet the distances were left unrestrained.
  • the hydrogen atoms on 099 (water) were not found from the difference map and left out of the structure solution.
  • the hydrogen parameters were added to the structure factor calculations but were not refined.
  • the shifts calculated in the final cycles of least squares refinement were all less than 0.2 of the corresponding standard deviations.
  • the final R-index was 3.58%.
  • a final difference Fourier revealed no missing or misplaced electron density. Of the residuals left, one is in a reasonable position for a proton bonded to 039 (carboxylic acid). This residual could be an additional occupancy position for the H98a proton (proton bonded to 039)
  • Anisotropic displacement parameters ( ⁇ 2 x 10 3 ) for Example 25.
  • the anisotropic displacement factor exponent takes the form: -2 ⁇ 2 [ h 2 a* 2 U 11 + ... + 2 h k a* b*U 12 ] U 11 U 22 U 33 U 23 U 13 U 12 C(1) 115(4) 49(2) 104(3) -20(2) -54(3) 31(2) C(2) 95(3) 52(2) 95(3) -12(2) -47(2) 23(2) O(3) 62(1) 36(1) 68(1) -3(1) -17(1) 6(1) C(4) 45(2) 35(2) 49(2) 1(1) 1(1) 0(1) C(5) 61(2) 43(2) 59(2) 4(2) -16(2) 9(2) C(6) 61(2) 43(2) 47(2) 2(1) -13(1) 0(2) C(7) 50(2) 36(2) 41(1) 6(1) 7(1) -2(1) C(8) 58(2) 44(2) 49(2) 9(1) -9(2) 7(2) C(9) 54(2) 43(2) 50(2) 2(1) -12(1) 3(1) C(10) 63(2) 40(2) 43(2) 6(1) 5(1) 6(1) C(11) 52(2) 27(1) 41
  • the packed rotor was oriented at the magic angle and spun at 15.0 kHz.
  • the 13 C solid state spectrum was collected using a proton decoupled cross-polarization magic angle spinning experiment (CPMAS).
  • the cross-polarization contact time was set to 2.0 ms.
  • a proton decoupling field of approximately 85 kHz was applied.
  • 1448 scans were collected with recycle delay of 14 seconds.
  • the carbon spectrum was referenced using an external standard of crystalline adamantane, setting its upfield resonance to 29.5 ppm.
  • the chemical shift data is dependent on the testing conditions (i.e. spinning speed and sample holder), reference material, and data processing parameters, among other factors.
  • the ss-NMR results are accurate to within about ⁇ 0.2 ppm.
  • the SSNMR 13 C CPMAS spectrum of Example 26 is shown in Figure 9 .
  • the peaks marked by asterisks in Figure 9 are spinning sidebands.
  • the SGLT2 functional assay was designed to detect the inhibition of methyl-alpha-D glucopyranoside (AMG - a non-metabolizable form of glucose) uptake via the SGLT2 transporter.
  • the SGLT2 transporter recovers glucose from the proximal tubules of the kidney; its inhibition results in sugar wasted in the urine.
  • the positive control compound, Phlorizin is a known inhibitor of glucose uptake for SGLT2 and was used for comparing the high percent effect of SGLT2 inhibition of the test compounds.
  • CHO-FlpIn Invitrogen, Carlsbad, CA cells stably expressing human SGLT2 (pcDNA5/FRT) were plated in Iso-TC 96 well plates (Perkin Elmer, Waltham, MA) at a density of 100,000 cells/well in 100 microL of growth media (1:1 F-12/DMEM media (Gibco, Carlsbad, CA), 10% FBS (Sigma, St. Louis MO), 1X Pen/Strep (Gibco, Carlsbad, CA), 600 microg/mL Hygromycin (Invitrogen, Carlsbad, CA)).
  • growth media 1:1 F-12/DMEM media (Gibco, Carlsbad, CA), 10% FBS (Sigma, St. Louis MO), 1X Pen/Strep (Gibco, Carlsbad, CA), 600 microg/mL Hygromycin (Invitrogen, Carlsbad, CA)).
  • test compounds Prior to treating with test compound, confluent cells were serum starved for 2 hours at 37°C in 1:1 F-12/DMEM media, replacing with fresh F-12/DMEM media after 1 hour.
  • Test compounds in dimethylsulfoxide (Sigma, St. Louis, MO) were diluted 100 fold in uptake buffer (140mM NaCl (Promega, Madison, WI), 2mM KCI (Teknova, Hollister, CA), 1mM CaCl 2 (Teknova, Hollister, CA), 1mM MgCl 2 (Teknova, Hollister, CA), and 10mM HEPES (Gibco, Carlsbad, CA) to cell plates pre-rinsed with uptake buffer.
  • uptake buffer 140mM NaCl (Promega, Madison, WI)
  • 2mM KCI Teknova, Hollister, CA
  • 1mM CaCl 2 Teknova, Hollister, CA
  • 1mM MgCl 2 Teknova, Hollister, CA
  • AMG 40 nCi AMG [U- 14 C] (Perkin Elmer, Waltham, MA) in unlabelled AMG (Aldrich, St. Louis, MO)) per well yielding a final concentration of 11.33 microM AMG.
  • Cell plates were then incubated for 3 hours at 37°C for AMG uptake. After incubation, cells were washed twice with ice cold wash buffer (uptake buffer containing 200 microM Phlorizin (Sigma), air dried and lysed in 30 microL of 200 mM NaOH and 1% SDS buffer on an orbital shaker.
  • Microscint 40 (Perkin Elmer, Waltham, MA) was added to the lysed cells (giving a final volume of 200 microL) and mixed by orbital shaking for 30 minutes. Plates were stored in the dark overnight and quantitated in the 1540 Microbeta Trilux (Wallac, Waltham, MA) using a normalized protocol for 14 C detection.
  • % Effect ZPE - T / ZPE - HPE x 100 %
  • ZPE the corrected counts per minute (CCPM) in control wells containing 0.5% DMSO
  • T the CCPM in wells containing test compound at various concentrations of the standard curve
  • HPE the high percent effect referring to the CCPM in control wells containing 10 microM Phlorizin.
  • IC 50 values were calculated using a dose response equation and are summarized for the compounds tested in Table 3.
  • Examples 1A and 4A were tested in rats to assess inhibition of glucose transport via urinary glucose excretion.
  • Dosing solutions were 0.03 mg/mL, 0.3 mg/mL, 0.9 mg/mL, 3 mg/mL, 9 mg/mL and 18 mg/mL for the 0.1 mg/kg, 1 mg/kg, 3 mg/kg, 10 mg/kg, 30 mg/kg and 60 mg/kg doses respectively.
  • Dosing volume was 1 mL/300 g of body weight for all doses.
  • Example 1A One group received a 10 mg/kg dose of Example 1A and others received 0.1, 1, 3, 10, 30 or 60 mg/kg dose of Example 4A.
  • the vehicle was 20% v/v PEG400 and 24% v/v hydroxypropyl beta cyclodextrin; HPBCD.
  • urine was collected for 24 hours.
  • Glucose concentration was measured in urine by UV absorbance spectrophotometry at 340 nm using a Roche Hitachi 917 spectrophotometer (Diamond Diagnostics, Holliston, MA). The total amount of glucose excreted in the urine was calculated as the product of urine concentration and urine volume using the formula below:
  • Examples 1A, 2A, 4A, 12A, and 14A were tested in rats to assess pharmacokinetic parameters including maximum concentration (Cmax), area under the plasma concentration time curve (AUC), clearance (CL), steady state volume of distribution (Vss), half life (t 1/2 ), and bioavailability (F).
  • Cmax maximum concentration
  • AUC area under the plasma concentration time curve
  • CL clearance
  • Vss steady state volume of distribution
  • t 1/2 half life
  • bioavailability F.
  • Male Sprague Dawley rats ⁇ 300 g
  • Rats received compound by intraveneous (IV) or oral gavage (PO) administration and the doses tested including vehicle to formulate dosing solutions are listed in Table 5.
  • 0.2 mL blood was sampled from the jugular vein at various timepoints (Table 5). Twenty microL aliquots of plasma samples and standards were subjected to protein precipitation with acetonitrile containing an internal standard. Samples were vortexed and centrifuged to obtain supernatant which was analyzed by LC-MS/MS. Analyst (Version 1.4.1) was used to measure peak areas and peak area ratios of analyte to internal standard were calculated.
  • LC-MS/MS conditions are as follows: Mass Spectrometer + Source Type was Sciex API 4000 - Turbo Spray; HPLC pump was Shimadzu; Autosampler was CTC PAL Autosampler; Injection volume was 3.0 to 10 microL; A gradient was used with mobile phase A: 10 mM ammonium acetate and 1% Isopropyl alcohol in water; B: Acetonitrile; Flowrate 0.300 mL per minute (Column 2.0 x 30 mm 5 microm LUNA C18 column (phenomenex)). Detection mode was negative.
  • a calibration curve was constructed from the peak area ratios of the standards to the internal standard by applying a weighted linear (1/x or 1/x2) regression.
  • the dynamic range of the standard curve was 5.00 ng/mL to 5000 ng/mL.

Abstract

Compounds of Formula (I) are described herein and the uses thereof for the treatment of diseases, conditions and/or disorders mediated by sodium-glucose transporter inhibitors (in particular, SGLT2 inhibitors).The compounds disclosed herein are useful for the prevention and treatment of obesity and its associated co-morbidities, inn particular type Il (type 2) diabetes.

Description

    FIELD OF THE INVENTION
  • The present invention relates to dioxa-bicyclo[3.2.1]octane-2,3,4-triol derivatives, crystal structures, pharmaceutical compositions and the uses thereof as sodium-glucose co-transporter (SGLT) inhibitors.
  • BACKGROUND
  • Obesity is a significant health problem due to its serious medical complications that include co-morbidities such as hypertension, insulin resistance, diabetes, coronary artery disease and heart failure (collectively referred to as Metabolic Syndrome). Obesity and its related co-morbidities continue to cause rising health issues in the developed world and are beginning to affect the developing world as well. The negative health consequences of obesity make it the second leading cause of preventable death in the United States and impart a significant economic and psychosocial effect on society. See, McGinnis M, Foege WH., "Actual Causes of Death in the United States," JAMA, 270, 2207-12 (1993). There is a need to identify and develop new medications that treat and/or prevent obesity and its associated co-morbidities, in particular type II (type 2) diabetes.
  • More recently, sodium-glucose co-transport (SGLT) inhibitors, particularly SGLT2 inhibitors, have been shown to block the reabsorption of glucose from the renal filtrate in the glomerulus thereby inducing glucose excretion in the urine. As excess glucose is excreted, there is a decrease in blood glucose level, decreased hepatic storage of glucose, decreased insulin secretion and, subsequently, decreased carbohydrate conversion to fat and, ultimately, reduced accumulated fat. Selective inhibition of SGLT2 is expected to normalize plasma glucose by enhancing glucose excretion. Consequently, SGLT2 inhibitiors provide an attractive means for the improvement of diabetic conditions without increasing body weight or the risk of hypoglycemia. See, Isaji, M., Current Opinion Investigational Drugs, 8(4), 285-292 (2007). For a general review of SGLT as a therapeutic target, see also Asano, T., et al., Drugs of the Future, 29(5), 461-466 (2004).
  • Representative examples of glycosides that have been shown to be useful for the treatment of NIDDM and obesity can be found in the following disclosures: U.S. Patent Nos. 6,515,117 ; 6,414,126 ; 7,101,856 ; 7,169,761 ; and 7,202,350 ; U.S. Publication Nos. US2002/0111315 ; US2002/0137903 ; US2004/0138439 ; US2005/0233988 ; US2006/0025349 ; US2006/0035841 ; and US2006/0632722 ; and PCT Publication Nos. WO01/027128 ; WO02/044192 ; WO02/088157 ; WO03/099836 ; WO04/087727 ; WO05/021566 ; WO05/085267 ; WO06/008038 ; WO06/002912 ; WO06/062224 ; WO07/000445 ; WO07/093610 ; and WO08/002824 . Spiroketal derivatives as SGLT2 inhibitors are disclosed in WO2008/013280 .
  • Certain glycosides are genotoxic and impact a cell's genetic material such that they may be potentially mutagenic or carcinogenic. Genotoxic materials may be detected using standard assays such as the In Vitro Mammalian Cell Micronuleus Test (MNvit), Organization for Economic Co-Operation and Development (OECD) Draft Test Guideline (Draft TG) 487 (2007); In vitro Mammalian Chromosomal Aberration Test, OECD TG 473 (1997); Bacterial Reverse Mutation Test, OECD TG 471 (1997); Mammalian Erythrocyte Micronucleus Test, OECD TG 474 (1997); or the like. Consequently, there still exists a need for a more effective and safe therapeutic treatment and/or prevention of obesity and its associated co-morbidities, in particular, Type 2 diabetes and related disorders.
  • SUMMARY
  • Compounds of Formula (A) and Formula (B) have been found to act as sodium-glucose cotransport (SGLT) inhibitors, in particular, SGLT2 inhibitors; therefore, may be used in the treatment of diseases mediated by such inhibition (e.g., diseases related to obesity, Type 2 diabetes, and obesity-related and diabetes-related co-morbidities). These compounds may be represented by Formulas (A) and (B) as shown below:
    Figure imgb0001
    wherein R1 is H, (C1-C4)alkyl, (C1-C4)alkoxy, Cl, F, cyano, fluoro-substituted (C1-C2)alkyl, (C1-C4)alkyl-SO2-, or (C3-C6)cycloalkyl; and
    R2 is (C1-C4)alkyl, (C1-C4)alkoxy, (C2-C4)alkynyl, 3-oxetanyloxy, 3-tetrahydrofuranyloxy, Cl, F, cyano, fluoro-substituted (C1-C2)alkyl, (C1-C4)alkyl-SO2-, (C3-C6)cycloalkyl, or a (C5-C6)heterocycle having 1 or 2 heteroatoms each independently selected from N, O, or S.
  • It is generally understood by those skilled in the art that various substituents may be added to the compounds of Formula (A) or Formula (B) so long as the substituent(s) selected does not adversely affect the pharmacological characteristics of the compound or adversely interfere with the use of the medicament.
  • Specific compounds of Formula (A) include: (1S,2S,3S,4R,5S)-1-hydroxymethyl-5-[3-(4-methoxy-benzyl)-4-methyl-phenyl]-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4R,5S)-5-[3-(4-ethoxy-benzyl)-4-methyl-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4R,5S)-5-[4-fluoro-3-(4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; 2-(4-methoxybenzyl)-4-((1S,2S,3S,4R,5S)-2,3,4-trihydroxy-1-(hydroxymethyl)-6,8-dioxa-bicyclo[3,2,1]oct-5-yl)benzonitrile; 2-(4-ethoxybenzyl)-4-((1S,2S,3S,4R,5S)-2,3,4-trihydroxy-1-(hydroxymethyl)-6,8-dioxa-bicyclo[3,2,1]oct-5-yl]benzonitrile; (1S,2S,3S,4R,5S)-5-[3-(4-ethoxy-benzyl)-4-fluorophenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4R,5S)-5-{4-fluoro-3-[4-(tetrahydro-furan-3-yloxy)-benzyl]-phenyl}-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4R,5S)-5-[3-(4-chlorobenzyl)-4-fluorophenyl]-1-hydroxymethyl-6,8-dioxabicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4R,5S)-5-{4-fluoro-3-[4-(oxetan-3-yloxy)-benzyl]-phenyl}-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; and (1S,2S,3S,4R,5S)-5-{4-chloro-3-[4-(oxetan-3-yloxy)-benzyl]-phenyl}-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol.
  • Specific compounds of Formula (B) include: (1S,2S,3S,4S,5S)-1-hydroxymethyl-5-[3-(4-methoxy-benzyl)-4-methyl-phenyl]-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4S,5S)-5-[3-(4-ethoxy-benzyl)-4-methyl-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4S,5S)-5-[4-chloro-3-(4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4S,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4S,5S)-5-[4-fluoro-3-(4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; (1S,2S,3S,4S,5S)-5-[3-(4-ethoxy-benzyl)-4-fluorophenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; and (1S,2S,3S,4S,5S)-5-[3-(4-chlorobenzyl)-4-fluorophenyl]-1-hydroxymethyl-6,8-dioxabicyclo[3.2.1]octane-2,3,4-triol.
  • A further aspect of the present invention is a crystal comprising a compound having the formula (4A):
    Figure imgb0002
  • Another aspect of the present invention is a pharmaceutical composition that comprises (1) a compound of the present invention, and (2) a pharmaceutically acceptable excipient, diluent, or carrier. Preferably, the composition comprises a therapeutically effective amount of a compound of the present invention. The composition may also contain at least one additional pharmaceutical agent (described herein). Preferred agents include anti-obesity agents and/or anti-diabetic agents (described herein below).
  • In yet another aspect of the present invention, a use of the compounds of the present invention in a method for treating a disease, disorder, or condition modulated by SGLT2 inhibition in animals is provided that includes the step of administering to an animal (preferably, a human) in need of such treatment a therapeutically effective amount of a compound of the present invention (or a pharmaceutical composition thereof). Diseases, conditions, and/or disorders modulated by SGLT2 inhibition include, e.g., Type II diabetes, diabetic nephropathy, insulin resistance syndrome, hyperglycemia, hyperinsulinemia, hyperlipidemia, impaired glucose tolerance, obesity (including weight control or weight maintenance), hypertension, and reducing the level of blood glucose.
  • Compounds of the present invention may be administered in combination with other pharmaceutical agents (in particular, anti-obesity and anti-diabetic agents described herein below). The combination therapy may be administered as (a) a single pharmaceutical composition which comprises a compound of the present invention, at least one additional pharmaceutical agent described herein and a pharmaceutically acceptable excipient, diluent, or carrier; or (b) two separate pharmaceutical compositions comprising (i) a first composition comprising a compound of the present invention and a pharmaceutically acceptable excipient, diluent, or carrier, and (ii) a second composition comprising at least one additional pharmaceutical agent described herein and a pharmaceutically acceptable excipient, diluent, or carrier. The pharmaceutical compositions may be administered simultaneously or sequentially and in any order.
  • It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention, as claimed.
  • BRIEF DESCRIPTION OF FIGURES
    • FIGURE 1 represents the refined crystal structure for the Example 8A compound which was plotted using the SHELXTL plotting package.
    • FIGURE 2 represents the refined crystal structure for the Example 9A compound which was plotted using the SHELXTL plotting package.
    • FIGURE 3 represents an observed powder X-ray diffraction pattern for Example 22: Example 18 Cocrystal of Example 4A compound and L-proline
    • FIGURE 4 represents an observed powder X-ray diffraction pattern for Example 22: Example 20 Cocrystal of Example 4A compound and L-pyroglutamic acid
    • FIGURE 5 represents an observed differential scanning calorimetry thermogram for Example 23: Example 18 Cocrystal of Example 4A compound and L-proline
    • FIGURE 6 represents an observed differential scanning calorimetry thermogram for Example 23: Example 20 Cocrystal of Example 4A compound and L-pyroglutamic acid
    • FIGURE 7 represents the refined crystal structure for the Example 24: Cocrystal of Example 4A compound and L-proline which was plotted using the SHELXTL plotting package
    • FIGURE 8 represents the refined crystal structure for the Example 25: Cocrystal of Example 4A compound and L-pyroglutamic acid which was plotted using the SHELXTL plotting package.
    • FIGURE 9 represents an observed 13C solid state nuclear magnetic resonance spectrum for Example 26: Cocrystal of Example 4A compound and L-pyroglutamic acid. The peaks marked by asterisks are spinning sidebands.
    DETAILED DESCRIPTION
  • The present invention may be understood even more readily by reference to the following detailed description of exemplary embodiments of the invention and the examples included therein.
  • Before the present compounds, compositions and methods are disclosed and described, it is to be understood that this invention is not limited to specific synthetic methods of making that may of course vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. The plural and singular should be treated as interchangeable, other than the indication of number:
    • As used herein, the term "alkyl" refers to a hydrocarbon radical of the general formula CnH2n+1. The alkane radical may be straight or branched. For example, the term "(C1-C6)alkyl" refers to a monovalent, straight, or branched aliphatic group containing 1 to 6 carbon atoms (e.g., methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, s-butyl, t-butyl, n-pentyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, neopentyl, 3,3-dimethylpropyl, hexyl, 2-methylpentyl, and the like). Similarly, the alkyl portion (i.e., alkyl moiety) of an alkoxy, acyl (e.g., alkanoyl), alkylamino, dialkylamino, alkylsulfonyl, and alkylthio group have the same definition as above. When indicated as being "optionally substituted", the alkane radical or alkyl moiety may be unsubstituted or substituted with one or more substituents (generally, one to three substituents except in the case of halogen substituents such as perchloro or perfluoroalkyls) independently selected from the group of substituents listed below in the definition for "substituted." "Halo-substituted alkyl" refers to an alkyl group substituted with one or more halogen atoms (e.g., fluoromethyl, difluoromethyl, trifluoromethyl, perfluoroethyl, 1,1-difluoroethyl and the like).
  • The term "cycloalkyl" refers to nonaromatic rings that are fully hydrogenated and may exist as a single ring, bicyclic ring or a spiro ring. Unless specified otherwise, the carbocyclic ring is generally a 3- to 8-membered ring. For example, cycloalkyl include groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, norbornyl (bicyclo[2.2.1]heptyl), bicyclo[2.2.2]octyl, and the like.
  • The term "heterocycle" refers to nonaromatic rings that are fully hydrogenated and may exist as a single ring, bicyclic ring or a spiral ring. Unless specified otherwise, the heterocyclic ring is generally a 3- to 6-membered ring containing 1 to 3 heteroatoms (preferably 1 or 2 heteroatoms) independently selected from sulfur, oxygen and/or nitrogen. Heterocyclic rings include groups such as epoxy, aziridinyl, tetrahydrofuranyl, pyrrolidinyl, N-methylpyrrolidinyl, piperidinyl, piperazinyl, pyrazolidinyl, 4H-pyranyl, morpholino, thiomorpholino, tetrahydrothienyl, tetrahydrothienyl 1,1-dioxide, and the like.
  • The phrase "therapeutically effective amount" means an amount of a compound of the present invention that (i) treats the particular disease, condition, or disorder, (ii) attenuates, ameliorates, or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein.
  • The term "animal" refers to humans (male or female), companion animals (e.g., dogs, cats and horses), food-source animals, zoo animals, marine animals, birds and other similar animal species. "Edible animals" refers to food-source animals such as cows, pigs, sheep and poultry.
  • The phrase "pharmaceutically acceptable" indicates that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
  • The terms "treating", "treat", or "treatment" embrace both preventative, i.e., prophylactic, and palliative treatment.
  • The terms "modulated" or "modulating", or "modulate(s)", as used herein, unless otherwise indicated, refers to the inhibition of the sodium-glucose transporter (in particular, SGLT2) with compounds of the present invention thereby partially or fully preventing glucose transport across the transporter.
  • The term "compounds of the present invention" (unless specifically identified otherwise) refer to compounds of Formula (A), Formula (B) and all pure and mixed stereoisomers (including diastereoisomers and enantiomers), tautomers and isotopically labeled compounds. Hydrates and solvates of the compounds of the present invention are considered compositions of the present invention, wherein the compound is in association with water or solvent, respectively. The compounds may also exist in one or more crystalline states, i.e. as co-crystals, polymorphs, or they may exist as amorphous solids. All such forms are encompassed by the claims.
  • In one embodiment, R1 is H, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, F, Cl, cyano, -CF3, cyclopropyl, or cyclobutyl. In another embodiment, R1 is H, methyl, ethyl, isopropyl, methoxy, ethoxy, F, Cl, cyano, -CF3, or cyclopropyl. In a further embodiment, R1 is H, methyl, ethyl, methoxy, ethoxy, F, Cl, cyano, -CF3, or cyclopropyl. In yet a further embodiment, R1 is methyl, ethyl, F, Cl, cyano, CF3, or cyclopropyl.
  • In one embodiment, R2 is methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, F, Cl, cyano, -CF3, -CF2CH3, ethynyl, 3-oxetanyloxy, 3-tetrahydrofuranyloxy, or cyclopropyl. In another embodiment, R2 is methyl, ethyl, isopropyl, methoxy, ethoxy, F, Cl, cyano, -CF3,-CF2CH3, ethynyl, 3-oxetanyloxy, 3-tetrahydrofuranyloxy, or cyclopropyl. In a further embodiment, R2 is methyl, ethyl, methoxy, ethoxy, F, Cl, cyano, -CF3, -CF2CH3, ethynyl, 3-oxetanyloxy, 3-tetrahydrofuranyloxy, or cyclopropyl. In yet a further embodiment, R2 is methoxy or ethoxy.
  • In one embodiment, the crystal comprises the compound 4A and L-proline or L-pyroglutamic acid.
  • In a further embodiment, the crystal has one or more of the following:
    1. a) space group of P2(1)2(1)2(1) and unit cell parameters substantially equal to the following:
      • a = 7.4907(10) Åα= 90°.
      • b = 12.8626(15) Å β= 90°.
      • c = 28.029(4) Å γ = 90°;
    2. b) a powder x-ray diffraction pattern comprising 2-theta values of (CuKα radiation, wavelength of 1.54056Å) 6.4 ± 0.2, 16.7 ± 0.2, 17.4 ± 0.2 and 21.1 ± 0.2;
    3. c) a solid state 13C NMR spectrum having peak positions at 16.5 ± 0.2, 131.1 ± 0.2, 158.7 ± 0.2, and 181.5 ± 0.2 ppm as determined on a 500MHz spectrometer relative to crystalline adamantine of 29.5 ppm; or
    4. d) a differential scanning calorimetry thermogram having an endotherm of about 142.5 ± 2°C.
  • In a further embodiment, the crystal is a co-crystal comprising the compound of formula (4A) and L-pyroglutamic acid in a 1:1 stochiometric ratio.
  • Compounds of the present invention may be synthesized by synthetic routes that include processes analogous to those well-known in the chemical arts, particularly in light of the description contained herein. The starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, WI) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v. 1-19, Wiley, New York (1967-1999 ed.), or Beilsteins Handbuch der organischen Chemie, 4, Aufl. ed. Springer-Verlag, Berlin, including supplements (also available via the Beilstein online database)).
  • For illustrative purposes, the reaction schemes depicted below provide potential routes for synthesizing the compounds of the present invention as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the inventive compounds. Although specific starting materials and reagents are depicted in the schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
  • In the preparation of compounds of the present invention, protection of remote functionality of intermediates may be necessary. The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods. A "hydroxy-protecting group" refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality. Suitable hydroxyl-protecting groups (O-Pg) include for example, allyl, acetyl (Ac), silyl (like trimethylsily (TMS) or tert-butyldimethylsilyl (TBS)), benzyl (Bn), para-methoxybenzyl (PMB), trityl (Tr), para-bromobenzoyl, para-nitrobenzoyl and the like (benzylidene for protection of 1,3-diols). The need for such protection is readily determined by one skilled in the art. For a general description of protecting groups and their use, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991.
  • Scheme 1 outlines the general procedures one could use to provide compounds of the present invention.
    Figure imgb0003
  • Allyl 2,3,4-tri-O-benzyl-D-glucopyranoside (I-a, where Pg1 is a benzyl group) can be prepared by procedures described by Shinya Hanashima, et al., in Bioorganic & Medicinal Chemistry, 9, 367 (2001); Patricia A. Gent et al. in Journal of the Chemical Society. Perkin 1, 1835 (1974); Hans Peter Wessel in the Journal of Carbohydrate Chemistry, 7, 263, (1988); or Yoko Yuasa, et al., in Organic Process Research & Development, 8, 405-407 (2004). In step 1 of Scheme 1, the hydroxymethylene group can be introduced onto the glycoside by means of a Swern oxidation followed by treatment with formaldehyde in the presence of an alkali metal hydroxide (e.g., sodium hydroxide). This is referred to as an aldol-Cannizzaro reaction. The Swern oxidation is described by Kanji Omura and Daniel Swern in Tetrahedron, 34, 1651 (1978). Modifications of this process known to those of skill in the art may also be used. For example, other oxidants, like stabilized 2-iodoxybenzoic acid described by Ozanne, A. et al. in Organic Letters, 5, 2903 (2003), as well as other oxidants known by those skilled in the art can also be used. The aldol Cannizzaro sequence has been described by Robert Schaffer in the Journal of The American Chemical Society, 81, 5452 (1959) and Amigues, E.J., et al., in Tetrahedron, 63, 10042 (2007).
  • In step 2 of Scheme 1, protecting groups (Pg2) can be added by treating intermediate (I-b) with the appropriate reagents and procedures for the particular protecting group desired. For example, p-methoxybenzyl (PMB) groups may be introduced by treatment of intermediate (I-b) with p-methoxybenzyl bromide or p-methoxybenzyl chloride in the presence of sodium hydride, potassium hydride, potassium tert-butoxide in a solvent like tetrahydrofuran, 1,2-dimethoxyethane or N,N-dimethylformamide (DMF). Conditions involving para-methoxybenzyltrichloroacetimidate in presence of a catalytic amount of acid (e.g., trifluoromethanesulfonic acid, methanesulfonic acid, or camphorsulfonic acid) in a solvent such as dichloromethane, heptane or hexanes can also be used. Benzyl (Bn) groups may be introduced by treatment of intermediate (I-b) with benzyl bromide or benzyl chloride in the presence of sodium hydride, potassium hydride, potassium tert-butoxide in a solvent like tetrahydrofuran, 1,2-dimethoxyethane or N,N-dimethylformamide. Conditions involving benzyltrichloroacetimidate in presence of a catalytic amount of acid (e.g., trifluoromethanesulfonic acid, methanesulfonic acid, or camphorsulfonic acid) in a solvent such as dichloromethane, heptane or hexanes can also be used.
  • In step 3 of Scheme 1, the allyl protection group is removed (e.g., by treatment with palladium chloride in methanol; cosolvent like dichloromethane may also be used; other conditions known by those skilled in the art could also be used, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991) to form the lactol (I-d).
  • In step 4 of Scheme 1, oxidation of the unprotected hydroxyl group to an oxo group (e.g., Swern oxidation) then forms the lactone (I-e).
  • In step 5 of Scheme 1, the lactone (I-e) is reacted with N,O-dimethyl hydroxylamine hydrochloride to form the corresponding Weinreb amide which may exist in equilibrium in a closed/opened form. (I-f/I-g). The "Weinreb amide" (I-g) can be made using procedures well known to those of skill in the art. See, Nahm, S., and S.M. Weinreb, Tetrahedron Letters, 22 (39), 3815-1818 (1981). For example, intermediate (I-f/I-g) can be prepared from the commercially available N,O-dimethylhydroxylamine hydrochloride and an activating agent (e.g., trimethylaluminum).
  • In step 6 of Scheme 1, the arylbenzyl group (Ar) is introduced using the desired organometallic reagent (e.g., organo lithium compound (ArLi) or organomagnesium compound (ArMgX)) in tetrahydrofuran (THF) at a temperature ranging from about -78°C to about 20°C followed by hydrolysis (upon standing in protic conditions) to the corresponding lactol (I-i) which may be in equilibrium with the corresponding ketone (I-h). The bridged ketal motif found in (A) and (B) can be prepared by removing the protecting groups (Pg2) using the appropriate reagents for the protecting groups employed. For example, the PMB protecting groups may be removed by treatment with trifluoroacetic acid in the presence of anisole and dichloromethane (DCM) at about 0°C to about 23°C (room temperature). The remaining protecting groups (Pg1) may then be removed using the appropriate chemistry for the particular protecting groups. For example, benzyl protecting groups may be removed by treating with formic acid in the presence of palladium (Pd black) in a protic solvent (e.g., ethanol/THF) at about room temperature to produce the final products (A) and (B). When R1 is CN, the use of a Lewis acid like boron trichloride at a temperature ranging from about -78°C to about room temperature in a solvent like dichloromethane or 1,2-dichloroethane may also be used to remove benzyl protective and/or para-methoxybenzyl protective groups.
  • When R1 is CN and R2 is (C1-C4)alkoxy in intermdediate (I-i) or in products (A) or (B), upon treatment with a Lewis acid such as boron trichloride or boron tribomide, partial to complete de-alkylation to the corresponding phenol may occur to lead to the corresponding compound (A) or (B) where R1 is CN and R2 is OH. If this occurs, the (C1-C4)alkoxy group may be re-introduced via selective alkylation using a (C1-C4) alkyl iodide under mildly basic conditions, for example, potassium carbonate in acetone at a temperature ranging from about room temperature to about 56 degrees Celsius.
  • When R1 and/or R2 is (C1-C4)alkyl-SO2- it is understood by one skilled in the art that the organometallic addition step 6 (Scheme 1) will be carried out on the corresponding (C1-C4)alkyl-S- containing organometallic reagent. The thio-alkyl is then oxidized at a later stage to the corresponding sulfone using conventional methods known by those skilled in the art.
  • The compounds of the present invention may be prepared as co-crystals using any suitable method. A representative scheme for preparing such co-crystals is described in Scheme 2.
    Figure imgb0004
  • In Scheme 2, wherein Me is methyl and Et is ethyl, in step 1,1-(5-bromo-2-chlorobenzyl)-4-ethoxybenzene is dissolved in 3:1, toluene: tetrahydrofuran followed by cooling the resulting solution to <-70°C. To this solution is added hexyllithium while maintaining the reaction at ≤-65°C followed by stirring for 1 hour. (3R,4S,5R,6R)-3,4,5-tris(trimethylsilyloxy)-6-((trimethylsilyloxy)methyl)-tetrahydropyran-2-one (II-a) is dissolved in toluene and the resulting solution is cooled to -15°C. This solution is then added to the-70°C aryllithium solution followed by stirring for 1 hour. A solution of methanesulfonic acid in methanol is then added followed by warming to room temperature and stirring for 16 to 24 hours. The reaction is deemed complete when the α-anomer level is ≤ 3%. The reaction is then basified by the addition of 5 M aqueous sodium hydroxide solution. The resulting salts are filtered off followed by concentration of the crude product solution. 2-methyltetrahydrofuran is added as a co-solvent and the organic phase is extracted twice with water. The organic phase is then concentrated to 4 volumes in toluene. This concentrate is then added to a 5:1, heptane: toluene solution causing precipitate to form. The solids are collected and dried under vacuum to afford a solid.
  • In step 2 of Scheme 2, to (II-b) in methylene chloride is added imidazole followed by cooling to 0°C and then addition of trimethylsilylchloride to give the persilylated product. The reaction is warmed to room temperature and quenched by the addition of water, and the organic phase is washed with water. This crude methylene chloride solution of (II-c) is dried over sodium sulfate and then taken on crude into the next step.
  • In step 3 of Scheme 2, the crude solution of (II-c) in methylene chloride is concentrated to low volume and then the solvent is exchanged to methanol. The methanol solution of (II-c) is cooled to 0°C, then 1 mol% of potassium carbonate is added as a solution in methanol followed by stirring for 5 hours. The reaction is then quenched by addition of 1 mol% acetic acid in methanol, followed by warming to room temperature, solvent exchange to ethyl acetate, and then filtration of the minor amount of inorganic solids. The crude ethyl acetate solution of (II-d) is taken directly into the next step.
  • In step 4 of Scheme 2, the crude solution of (II-d) is concentrated to low volume, then diluted with methylene chloride and dimethylsulfoxide. Triethylamine is added followed by cooling to 10°C and then sulfur trioxide pyridine complex is added in 3 portions as a solid at 10 minute intervals. The reaction is stirred an additional 3 hours at 10°C before quenching with water and warming to room temperature. The phases are separated followed by washing the methylene chloride layer with aqueous ammonium chloride. The crude methylene chloride solution of (II-e) is taken directly into the next step.
  • In step 5 of Scheme 2, the crude solution of (II-e) is concentrated to low volume and then the solvent is exchanged to ethanol. Thirty equivalents of aqueous formaldehyde is added followed by warming to 55°C. An aqueous solution of 2 equivalents of potassium phosphate, tribasic is added followed by stirring for 24 hours at 55°C. The reaction temperature is then raised to 70°C for an additional 12 hours. The reaction is cooled to room temperature, diluted with tert-butyl methyl ether and brine. The phases are separated followed by solvent exchange of the organic phase to ethyl acetate. The ethyl acetate phase is washed with brine and concentrated to low volume. The crude concentrate is then purified by silica gel flash chromatography eluting with 5% methanol, 95% toluene. Product containing fractions are combined and concentrated to low volume. Methanol is added followed by stirring until precipitation occurs. The suspension is cooled and the solids are collected and rinsed with heptane followed by drying. Product (II-f) is isolated as a solid.
  • In step 6 of Scheme 2, compound (II-f) is dissolved in 5 volumes of methylene chloride followed by the addition of 1mol% SiliaBond® tosic acid and stirring for 18 hours at room temperature. The acid catalyst is filtered off and the methylene chloride solution of (II-g) is taken directly into the next step co-crystallization procedure.
  • In step 7 of Scheme 2, the methylene chloride solution of (II-g) is concentrated and then the solvent is exchanged to 2-propanol. Water is added followed by warming to 55°C. An aqueous solution of L-pyroglutamic acid is added followed by cooling the resulting solution to room temperature. The solution is then seeded and granulated for 18 hours. After cooling, the solids are collected and rinsed with heptane followed by drying. Product (II-h) is isolated as a solid.
  • An alternative synthesis route for compounds (A) of the present invention is depicted in Scheme 3 and described below.
    Figure imgb0005
  • The synthesis of (III-a), where R3 is an alkyl or fluoro substituted alkyl (except for the carbon adjacent to the oxygen atom) can be prepared in a similar way as described in step 1 of Scheme 2. In step 1 of Scheme 3, the primary hydroxyl group is selectively protected by an appropriate protective group. For example, a trityl group (Pg3 = Tr) can be introduced by treatment of intermediate (III-a) with chlorotriphenylmethane in presence of a base like pyridine in a solvent like toluene, tetrahydrofuran or dichloromethane at a temperature ranging from about 0 degrees Celsius to about room temperature. Additional examples of such protective groups and experimental conditions are known by those skilled in the art and can be found in T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991.
  • In step 2 of Scheme 3, the secondary hydroxyl groups can be protected by the appropriate protecting groups. For example, benzyl groups (Pg4 is Bn) can be introduced by treatment of intermediate (III-b) with benzyl bromide or benzyl chloride in the presence of sodium hydride, potassium hydride, potassium tert-butoxide in a solvent like tetrahydrofuran, 1,2-dimethoxyethane or N,N-dimethylformamide at a temperature ranging from about 0 degrees Celsius to about 80 degrees Celsius. Acetyl or benzoyl groups (Pg4 = Ac or Bz) may be introduced by treatment of intermediate (III-b) with acetyl chloride, acetyl bromide or acetic anhydride or benzoyl chloride or benzoic anhydride in the presence of a base like triethylamine, N,N-diisopropylethylamine or 4-(dimethylamino)pyridine in a solvent like tetrahydrofuran, 1,2-dimethoxyethane or dichloromethane at a temperature ranging from about 0 degrees Celsius to about 80 degrees Celsius.
  • In step 3 of Scheme 3, the primary hydroxyl group is deprotected to lead to intermediate (III-d). When Pg3 is Tr, intermediate (III-c) is treated in the presence of an acid like para-toluenesulfonic acid in a alcoholic solvent like methanol at a temperature ranging from about -20 degrees Celsius to about room temperature to provide intermediate (III-d). Cosolvents like chloroform may be used.
  • In step 4 of Scheme 3, a hydroxymethylene group is introduced through a process similar to the one already described in Scheme 1 (step 1) and Scheme 2 (steps 4 and 5). Other sources of formaldehyde, like paraformaldehyde in a solvent like ethanol at a temperature ranging from about room temperature to about 70 degrees Celsius in the presence of an alkali metal alkoxide can also be used in this step. When Pg4 is Bn, this step provides intermediate (III-e) and when Pg4 is Ac or Bz, this step provides intermediate (III-f).
  • In step 5 of Scheme 3, intermediate (III-e) is treated with an acid like trifluoroacetic acid or an acidic resin in a solvent like dichloromethane at a temperature ranging from about -10 degrees Celsius to about room temperature to produce intermediate (III-g).
  • In step 6 of Scheme 3, the remaining protecting groups (Pg4) may then be removed using the appropriate chemistry for the particular protecting groups. For example, benzyl protecting groups may be removed by treating with formic acid in the presence of palladium (Pd black) in a protic solvent (e.g., ethanol/THF) at about room temperature to produce the final product (A).
  • In step 7 of Scheme 3, intermediate (III-f) is treated with an acid like trifluoroacetic acid or an acidic resin in a solvent like dichloromethane at a temperature ranging from about -10 degrees Celsius to about room temperature to produce the final product (A).
  • Another alternative scheme for synthesizing product (A) is depicted in Scheme 4 and described below.
    Figure imgb0006
  • In step 1 of Scheme 4, intermediate (III-a) is treated with the appropriate arylsulfonyl chloride R4SO2Cl or arylsulfonic anhydride R4S(O)2OS(O)2R4 (wherein R4 is an optionally substituted aryl group, such as found in the arylsulfonyl chlorides 4-methyl-benzenesulfonyl chloride, 4-nitro-benzenesulfonyl chloride, 4-fluoro-benzenesulfonyl chloride, 2,6-dichloro-benzenesulfonyl chloride, 4-fluoro-2-methyl-benzenesulfonyl chloride, and 2,4,6-trichloro-benzenesulfonyl chloride, and in the arylsulfonic anhydride, p-toluenesulfonic anhydride) in presence of a base like pyridine, triethylamine, N,N-diisopropylethylamine in a solvent like tetrahydrofuran, 2-methyltetrahydrofuran at a temperature ranging from about -20 degrees Celsius to about room temperature. Some Lewis acids like zinc(II) bromide may be used as additives.
  • In step 2 of Scheme 4, intermediate (IV-a) is submitted to a Kornblum-type oxidation (see, Kornblum, N., et al., Journal of The American Chemical Society, 81, 4113 (1959)) to produce the corresponding aldehyde which may exist in equilibrium with the corresponding hydrate and/or hemiacetal form. For example intermediate (IV-a) is treated in the presence of a base like pyridine, 2,6-lutidine, 2,4,6-collidine, N,N-diisopropylethylamine, 4-(dimethylamino)pyridine in a solvent like dimethyl sulfoxide at a temperature ranging from about room temperature to about 150 degrees Celsius. The aldehyde intermediate produced is then submitted to the aldol/Cannizzaro conditions described for step 1 (Scheme 1) and step 5 (Scheme 2) to produce intermediate (IV-b).
  • In step 3 of Scheme 4, intermediate (IV-b) is treated with an acid like trifluoroacetic acid or an acidic resin in a solvent like dichloromethane at a temperature ranging from about -10 degrees Celsius to about room temperature to produce the final product (A).
  • When R2 is (C2-C4)alkynyl the process may be performed using Scheme 5, wherein R6 is H or (C1-C2)alkyl.
    Figure imgb0007
    Figure imgb0008
  • In step 1 of Scheme 5, which provides intermediate (V-i), the organometallic addition step is carried out in a similar way to the one described in Scheme1, step 6, using the organometallic reagent derived from (V-a), where Pg5 is a suitable protective group for the hydroxyl group. For instance Pg5 can be a tert-butyldimethylsilyl group (TBS) (see US2007/0054867 for preparation of for instance {4-[(5-bromo-2-chloro-phenyl)-methyl]-phenoxy}-tert-butyl-dimethyl-silane).
  • In step 2 of Scheme 5, when Pg2 = PMB, intermediate (V-i) is treated with an acid like trifluoroacetic acid, methanesulfonic acid or an acidic resin in presence of anisole in a solvent like dichloromethane at a temperature ranging from about -10 degrees Celsius to about room temperature to produce intermediate (V-j).
  • In step 3 of Scheme 5, protecting groups (Pg5) and (Pg1) can be removed to provide (V-k). Typically (Pg5) is TBS and Pg1 is Bn. In this circumstance, the protecting groups are removed by sequential treatment of (V-j) with 1) tetrabutylammonium fluoride in a solvent like tetrahydrofuran or 2-methyltetrahydrofuran at a temperature ranging from 0 degrees Celsius to about 40 degrees Celsius and 2) treatment with formic acid in the presence of palladium (Pd black) in a protic solvent (e.g., ethanol/THF) at about room temperature. In this sequence, the order of the 2 reactions is interchangeable.
  • In step 4 of Scheme 5, intermediate (V-k) is treated with N,N-bis-(trifluoromethanesulfonyl)-aniline in presence of a base like triethylamine or 4-dimethyaminopyridine in a solvent like dichloromethane or 1,2-dichloroethane at a temperature ranging from 0 degrees Celsius to about 40 degrees Celsius to produce intermediate (V-I).
  • In step 5 of Scheme 5, intermediate (V-I) is subjected to a Sonogashira-type reaction (see, Sonogashira, K. Coupling Reactions Between sp2 and sp Carbon Centers. In Comprehensive Organic Synthesis (eds. Trost, B. M., Fleming, I.), 3, 521-549, (Pergamon, Oxford, 1991)). For instance (V-I) is treated with the appropriate terminal alkyne HCCR6 in presence of copper(I) iodide, a catalyst like bis-(triphenylphosphine)-palladium dichloride or tetrakis(triphenylphosphine)palladium(0) in presence of a base like triethylamine or N,N-diisopropylethylamine in a solvent like N,N-dimethylformamide at a temperature ranging from about room temperature to about 120 degrees Celsius to produce the desired product (A) and (B). When R6 is H, it is more convenient to use trimethylsilylacetylene. In this case the crude material obtained from the reaction described above is treated with a base like potassium carbonate in an alcoholic solvent like MeOH at about room temperature to produce after classical work-up known by those skilled in the art the desired product (A) and (B) where R2 is -CCH.
  • One skilled in the art would understand that the chemistry described above in schemes 1 to 5, represents different ways of accessing intermediate (V-k). In turn, particularly when R1 is Cl, (V-k) can be treated with an alkylating agent of choice under classical conditions to selectively alkylate the phenol group to produce (A) (and (B) in schemes 1 and 5) where R2 is (C1-C4)alkoxy.
  • The compounds of the present invention contain asymmetric or chiral centers, and, therefore, exist in different stereoisomeric forms. Unless specified otherwise, it is intended that all stereoisomeric forms of the compounds of the present invention as well as mixtures thereof, including racemic mixtures, form part of the present invention. In addition, the present invention embraces all geometric and positional isomers. For example, if a compound of the present invention incorporates a double bond or a fused ring, both the cis- and trans- forms, as well as mixtures, are embraced within the scope of the invention.
  • Diastereomeric mixtures can be separated into their individual diastereoisomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization, distillation, sublimation. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereoisomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers. Also, some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention. Enantiomers can also be separated by use of a chiral HPLC (high pressure liquid chromatography) column.
  • It is also possible that the intermediates and compounds of the present invention may exist in different tautomeric forms, and all such forms are embraced within the scope of the invention. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies which are interconvertible via a low energy barrier. For example, proton tautomers (also known as prototropic tautomers) include interconversions via migration of a proton, such as keto-enol and imine-enamine isomerizations. A specific example of a proton tautomer is the imidazole moiety where the proton may migrate between the two ring nitrogens. Valence tautomers include interconversions by reorganization of some of the bonding electrons. The equilibrium between closed and opened form of some intermediates (and/or mixtures of intermediates) is reminiscent of the process of mutarotation involving aldoses, known by those skilled in the art.
  • The present invention also embraces isotopically-labeled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, iodine, and chlorine, such as 2H, 3H, 11C, 13C, 14C, 13N, 15N, 15O, 17O, 18O, 31P, 32P, 35S, 18F, 123I, 125I and 36Cl, respectively.
  • Certain isotopically-labeled compounds of the present invention (e.g., those labeled with 3H and 14C) are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e., 3H) and carbon-14 (i.e., 14C) isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Positron emitting isotopes such as 15O, 13N, 11C, and 18F are useful for positron emission tomography (PET) studies to examine substrate occupancy. Isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples herein below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • Compounds of the present invention are useful for treating diseases, conditions and/or disorders modulated by the inhibition of the sodium-glucose transporters (in particular SGLT2); therefore, another embodiment of the present invention is a pharmaceutical composition comprising a therapeutically effective amount of a compound of the present invention and a pharmaceutically acceptable excipient, diluent or carrier. The compounds of the present invention (including the compositions and processes used therein) may also be used in the manufacture of a medicament for the therapeutic applications described herein.
  • A typical formulation is prepared by mixing a compound of the present invention and a carrier, diluent or excipient. Suitable carriers, diluents and excipients are well known to those skilled in the art and include materials such as carbohydrates, waxes, water soluble and/or swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water, and the like. The particular carrier, diluent or excipient used will depend upon the means and purpose for which the compound of the present invention is being applied. Solvents are generally selected based on solvents recognized by persons skilled in the art as safe (GRAS) to be administered to a mammal. In general, safe solvents are non-toxic aqueous solvents such as water and other non-toxic solvents that are soluble or miscible in water. Suitable aqueous solvents include water, ethanol, propylene glycol, polyethylene glycols (e.g., PEG400, PEG300), etc. and mixtures thereof. The formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
  • The formulations may be prepared using conventional dissolution and mixing procedures. For example, the bulk drug substance (i.e., compound of the present invention or stabilized form of the compound (e.g., complex with a cyclodextrin derivative or other known complexation agent)) is dissolved in a suitable solvent in the presence of one or more of the excipients described above. The compound of the present invention is typically formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to give the patient an elegant and easily handleable product.
  • The pharmaceutical compositions also include solvates and hydrates of the compounds of Formula (I). The term "solvate" refers to a molecular complex of a compound represented by Formula (I) (including pharmaceutically acceptable salts thereof) with one or more solvent molecules. Such solvent molecules are those commonly used in the pharmaceutical art, which are known to be innocuous to the recipient, e.g., water, ethanol, ethylene glycol, and the like, The term "hydrate" refers to the complex where the solvent molecule is water. The solvates and/or hydrates preferably exist in crystalline form. Other solvents may be used as intermediate solvates in the preparation of more desirable solvates, such as methanol, methyl t-butyl ether, ethyl acetate, methyl acetate, (S)-propylene glycol, (R)-propylene glycol, 1,4-butyne-diol, and the like. The crystalline forms may also exist as complexes with other innocuous small molecules, such as L-phenylalanine, L-proline, L-pyroglutamic acid and the like, as co-crystals or solvates or hydrates of the co-crystalline material. The solvates, hydrates and co-crystalline compounds may be prepared using procedures described in PCT Publication No. WO 08/002824 , incorporated herein by reference, or other procedures well-known to those of skill in the art.
  • The pharmaceutical composition (or formulation) for application may be packaged in a variety of ways depending upon the method used for administering the drug. Generally, an article for distribution includes a container having deposited therein the pharmaceutical formulation in an appropriate form. Suitable containers are well-known to those skilled in the art and include materials such as bottles (plastic and glass), sachets, ampoules, plastic bags, metal cylinders, and the like. The container may also include a tamper-proof assemblage to prevent indiscreet access to the contents of the package. In addition, the container has deposited thereon a label that describes the contents of the container. The label may also include appropriate warnings.
  • The present invention further provides a use of the compounds of the present invention in a method of treating diseases, conditions and/or disorders modulated by the inhibition of sodium-glucose transporters in an animal that includes administering to an animal in need of such treatment a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition comprising an effective amount of a compound of the present invention and a pharmaceutically acceptable excipient, diluent, or carrier. The method is particularly useful for treating diseases, conditions and/or disorders that benefit from the inhibition of SGLT2.
  • One aspect of the present invention is the treatment of obesity, and obesity-related disorders (e.g., overweight, weight gain, or weight maintenance).
  • Obesity and overweight are generally defined by body mass index (BMI), which is correlated with total body fat and estimates the relative risk of disease. BMI is calculated by weight in kilograms divided by height in meters squared (kg/m2). Overweight is typically defined as a BMI of 25-29.9 kg/m2, and obesity is typically defined as a BMI of 30 kg/m2. See, e.g., National Heart, Lung, and Blood Institute, Clinical Guidelines on the Identification, Evaluation, and Treatment of Overweight and Obesity in Adults, The Evidence Report, Washington, DC: U.S. Department of Health and Human Services, NIH publication no. 98-4083 (1998).
  • Another aspect of the present invention is for the treatment or delaying the progression or onset of diabetes or diabetes-related disorders including Type 1 (insulin-dependent diabetes mellitus, also referred to as "IDDM") and Type 2 (noninsulin-dependent diabetes mellitus, also referred to as "NIDDM") diabetes, impaired glucose tolerance, delayed wound healing, hyperinsulinemia, elevated blood levels of fatty acids, hyperlipidemia, hypertriglyceridemia, Syndrome X, increased high density lipoprotein levels, insulin resistance, hyperglycemia, and diabetic complications (such as atherosclerosis, coronary heart disease, stroke, peripheral vascular disease, nephropathy, hypertension, neuropathy, and retinopathy).
  • In yet another aspect of the present invention is the treatment of obesity co-morbidities, such as metabolic syndrome. Metabolic syndrome includes diseases, conditions or disorders such as dyslipidemia, hypertension, insulin resistance, diabetes (e.g., Type 2 diabetes), coronary artery disease and heart failure. For more detailed information on Metabolic Syndrome, see, e.g., Zimmet, P.Z., et al., "The Metabolic Syndrome: Perhaps an Etiologic Mystery but Far From a Myth - Where Does the International Diabetes Federation Stand?," Diabetes & Endocrinology, 7(2), (2005); and Alberti, K.G., et al., "The Metabolic Syndrome - A New Worldwide Definition," Lancet, 366, 1059-62 (2005).
  • Preferably, administration of the compounds of the present invention provides a statistically significant (p<0.05) reduction in at least one cardiovascular disease risk factor, such as lowering of plasma leptin, C-reactive protein (CRP) and/or cholesterol, as compared to a vehicle control containing no drug. The administration of compounds of the present invention may also provide a statistically significant (p<0.05) reduction in glucose serum levels.
  • For a normal adult human having a body weight of about 100 kg, a dosage in the range of from about 0.001 mg to about 10 mg per kilogram body weight is typically sufficient, preferably from about 0.01 mg/kg to about 5.0 mg/kg, more preferably from about 0.01 mg/kg to about 1 mg/kg. However, some variability in the general dosage range may be required depending upon the age and weight of the subject being treated, the intended route of administration, the particular compound being administered and the like. The determination of dosage ranges and optimal dosages for a particular patient is well within the ability of one of ordinary skill in the art having the benefit of the instant disclosure. It is also noted that the compounds of the present invention can be used in sustained release, controlled release, and delayed release formulations, which forms are also well known to one of ordinary skill in the art.
  • The compounds of this invention may also be used in conjunction with other pharmaceutical agents for the treatment of the diseases, conditions and/or disorders described herein. Therefore, methods of treatment that include administering compounds of the present invention in combination with other pharmaceutical agents are also provided. Suitable pharmaceutical agents that may be used in combination with the compounds of the present invention include anti-obesity agents (including appetite suppressants), anti-diabetic agents, anti-hyperglycemic agents, lipid lowering agents, anti-inflammatory agents and anti-hypertensive agents.
  • Suitable anti-obesity agents include cannabinoid-1 (CB-1) antagonists (such as rimonabant), 11β-hydroxy steroid dehydrogenase-1 (11β-HSD type 1) inhibitors, stearoyl-CoA desaturase-1 (SCD-1) inhibitor, MCR-4 agonists, cholecystokinin-A (CCK-A) agonists, monoamine reuptake inhibitors (such as sibutramine), sympathomimetic agents, β3 adrenergic agonists, dopamine agonists (such as bromocriptine), melanocyte-stimulating hormone analogs, 5HT2c agonists, melanin concentrating hormone antagonists, leptin (the OB protein), leptin analogs, leptin agonists, galanin antagonists, lipase inhibitors (such as tetrahydrolipstatin, i.e. orlistat), anorectic agents (such as a bombesin agonist), neuropeptide-Y antagonists (e.g., NPY Y5 antagonists), PYY3-36 (including analogs thereof), thyromimetic agents, dehydroepiandrosterone or an analog thereof, glucocorticoid agonists or antagonists, orexin antagonists, glucagon-like peptide-1 agonists, ciliary neurotrophic factors (such as Axokine™ available from Regeneron Pharmaceuticals, Inc., Tarrytown, NY and Procter & Gamble Company, Cincinnati, OH), human agouti-related protein (AGRP) inhibitors, ghrelin antagonists, histamine 3 antagonists or inverse agonists, neuromedin U agonists, MTP/ApoB inhibitors (e.g., gut-selective MTP inhibitors, such as dirlotapide), opioid antagonist, orexin antagonist, and the like.
  • Preferred anti-obesity agents for use in the combination aspects of the present invention include CB-1 antagonists (e.g., rimonabant, taranabant, surinabant, otenabant, SLV319 (CAS No. 464213-10-3) and AVE1625 (CAS No. 358970-97-5)), gut-selective MTP inhibitors (e.g., dirlotapide, mitratapide and implitapide, R56918 (CAS No. 403987) and CAS No. 913541-47-6), CCKa agonists (e.g., N-benzyl-2-[4-(1H-indol-3-ylmethyl)-5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-tetraaza-benzo[e]azulen-6-yl]-N-isopropyl-acetamide described in PCT Publication No. WO 2005/116034 or US Publication No. 2005-0267100 A1 ), 5HT2c agonists (e.g., lorcaserin), MCR4 agonist (e.g., compounds described in US 6,818,658 ), lipase inhibitor (e.g., Cetilistat), PYY3-36 (as used herein "PYY3-36" includes analogs, such as peglated PYY3-36 e.g., those described in US Publication 2006/0178501 ), opioid antagonists (e.g., naltrexone), oleoyl-estrone (CAS No. 180003-17-2), obinepitide (TM30338), pramlintide (Symlin®), tesofensine (NS2330), leptin, liraglutide, bromocriptine, orlistat, exenatide (Byetta®), AOD-9604 (CAS No. 221231-10-3) and sibutramine. Preferably, compounds of the present invention and combination therapies are administered in conjunction with exercise and a sensible diet.
  • Suitable anti-diabetic agents include an acetyl-CoA carboxylase-2 (ACC-2) inhibitor, a phosphodiesterase (PDE)-10 inhibitor, a diacylglycerol acyltransferase (DGAT) 1 or 2 inhibitor, a sulfonylurea (e.g., acetohexamide, chlorpropamide, diabinese, glibenclamide, glipizide, glyburide, glimepiride, gliclazide, glipentide, gliquidone, glisolamide, tolazamide, and tolbutamide), a meglitinide, an α-amylase inhibitor (e.g., tendamistat, trestatin and AL-3688), an α-glucoside hydrolase inhibitor (e.g., acarbose), an α-glucosidase inhibitor (e.g., adiposine, camiglibose, emiglitate, miglitol, voglibose, pradimicin-Q, and salbostatin), a PPARγ agonist (e.g., balaglitazone, ciglitazone, darglitazone, englitazone, isaglitazone, pioglitazone, rosiglitazone and troglitazone), a PPAR α/γ agonist (e.g., CLX-0940, GW-1536, GW-1929, GW-2433, KRP-297, L-796449, LR-90, MK-0767 and SB-219994), a biguanide (e.g., metformin), a glucagon-like peptide 1 (GLP-1) agonist (e.g., exendin-3 and exendin-4), a protein tyrosine phosphatase-1B (PTP-1 B) inhibitor (e.g., trodusquemine, hyrtiosal extract, and compounds disclosed by Zhang, S., et al., Drug Discovery Today, 12(9/10), 373-381 (2007)), SIRT-1 inhibitor (e.g., reservatrol), a dipeptidyl peptidease IV (DPP-IV) inhibitor (e.g., sitagliptin, vildagliptin, alogliptin and saxagliptin), an insulin secreatagogue, a fatty acid oxidation inhibitor, an A2 antagonist, a c-jun amino-terminal kinase (JNK) inhibitor, insulin, an insulin mimetic, a glycogen phosphorylase inhibitor, a VPAC2 receptor agonist and a glucokinase activator. Preferred anti-diabetic agents are metformin and DPP-IV inhibitors (e.g., sitagliptin, vildagliptin, alogliptin and saxagliptin).
  • Suitable anti-inflammatory agents include genital tract/urinary tract infection preventatives and treatments. Exemplary agents include cranberries (i.e. Vaccinium macrocarpon) and cranberry derivatives such as cranberry juice, cranberry extracts or flavonols of cranberries. Cranberry extracts may include one or more flavonols (i.e. anthocyanins and proanthocyanidins) or a purified cranberry flavonol compound, including myricetin-3-β-xylopyranoside, quercetin-3-β-glucoside, quercetin-3-α-arabinopyranoside, 3'-methoxyquercetin-3-α-xylopyranoside, quercetin-3-O-(6"-p-coumaroyl)-β-galactoside, quercetin-3-O-(6"-benzoyl)-β-galactoside, and/or quercetin-3-α-arabinofuranoside.
  • Embodiments of the present invention are illustrated by the following Examples. It is to be understood, however, that the embodiments of the invention are not limited to the specific details of these Examples, as other variations thereof will be known, or apparent in light of the instant disclosure, to one of ordinary skill in the art.
  • EXAMPLES
  • Unless specified otherwise, starting materials are generally available from commercial sources such as Aldrich Chemicals Co. (Milwaukee, WI), Lancaster Synthesis, Inc. (Windham, NH), Acros Organics (Fairlawn, NJ), Maybridge Chemical Company, Ltd. (Cornwall, England), Tyger Scientific (Princeton, NJ), AstraZeneca Pharmaceuticals (London, England), and Accela ChemBio (San Diego, CA).
  • General Experimental Procedures
  • NMR spectra were recorded on a Varian Unity™ 400 (available from Varian Inc., Palo Alto, CA) at room temperature at 400 MHz for proton. Chemical shifts are expressed in parts per million (delta) relative to residual solvent as an internal reference. The peak shapes are denoted as follows: s, singlet; d, doublet; dd, doublet of doublet; t, triplet; q, quartet; m, multiplet; bs or br.s., broad singlet; 2s, two singlets; br.d., broad doublet. Electrospray ionization mass spectra (ES) were obtained on a Waters™ ZMD instrument (carrier gas: nitrogen; solvent A: water /0.01% formic acid, solvent B: acetonitrile/0.005% formic acid; available from Waters Corp., Milford, MA). High resolution mass spectra (HRMS) were obtained on an Agilent™ Model 6210 time of flight. Where the intensity of single chlorine or single bromine-containing ions are described, the expected intensity ratio was observed (approximately 3:1 for 35Cl/37Cl-containing ions and 1:1 for 79Br/81Br-containing ions) and the intensity of only the lower mass ion is given. In some cases only representative 1H NMR peaks are given.
  • Column chromatography was performed with either Baker™ silica gel (40 microm; J.T. Baker, Phillipsburg, NJ) or Silica Gel 50 (EM Sciences™, Gibbstown, NJ) in glass columns or in Flash 40 Biotage™ columns (ISC, Inc., Shelton, CT). MPLC (medium pressure liquid chromatography) was performed using a Biotage™ SP purification system or a Combiflash® Companion® from Teledyne™ Isco™; Biotage™ SNAP cartridge KPsil or Redisep Rf silica (from Teledyne™ Isco™) under low nitrogen pressure were used. HPLC (high pressure liquid chromatography) was performed using a Shimadzu™ 10A LC-UV or a Agilent™ 1100 preparatory HPLC.
    Except where otherwise noted, all reactions were run under an inert atmosphere of nitrogen gas using anhydrous solvents. Also, except where otherwise noted, all reactions were run at room temperature (~23°C).
    When doing TLC (thin layer chromatography), Rf is defined as the ratio of the distance traveled by the compound divided by the distance traveled by the eluent. Rt (retention time).
  • Starting Materials
  • Generally, any of the following starting materials can be prepared using the procedures described in Schemes 7 or 8 of US Publication No. 2008/0132563 , or alternatively, Schemes 2, 3 or 8 of US Publication No. 2007/0259821 . More specifically, the following starting materials used in the following Examples can be prepared using the procedures described in the corresponding references or purchased from the corresponding vendor.
  • 4-Bromo-2-(4-methoxy-benzyl)-1-methyl-benzene can be prepared by the procedures described in Example 8 of PCT Publication No. WO 01/027128 .
  • 4-Bromo-2-(4-ethoxy-benzyl)-1-methyl-benzene can be prepared by the procedures described in Preparation Example 17 of US2008/0132563 .
  • 4-Bromo-1-chloro-2-(4-methoxy-benzyl)-benzene can be prepared by the procedures described in Preparation Example 19 of US2008/0132563 or Example V of US2007/0259821 .
  • 4-Bromo-1-chloro-2-(4-ethoxy-benzyl)-benzene may be purchased from Shanghai Haoyuan Chemexpress Co., Ltd., Shanghai, People's Republic of China.
  • 4-Bromo-2-(4-methoxy-benzyl)-benzonitrile can be prepared by the procedures described in Example XXII of US2007/0259821 .
  • The following starting materials were prepared as described below.
  • Preparation of 4-bromo-1-fluoro-2-(4-methoxy-benzyl)-benzene:
  • Oxalyl chloride (11.0 mL, 126 mmol) was added dropwise to a well stirred suspension of 5-bromo-2-fluoro-benzoic acid (25.0 g, 114 mmol) in dichloromethane (150 mL) and N,N-dimethylformamide (1.5 mL) at 0°C. The resulting mixture was allowed to gradually warm to room temperature. After 18 hours, the solid had gone into solution. The resulting light orange solution was concentrated under reduced pressure and was chased two times with diethyl ether to afford 5-bromo-2-fluoro-benzoyl chloride (27.0 g, quantitative yield) as a pale orange oil.
  • To a solution of 5-bromo-2-fluoro-benzoyl chloride (27.0 g, 114 mmol) and anisole (12.9 g, 13.0 mL, 119 mmol) in dichloromethane (150 mL) at 0°C was added aluminum trichloride (16.2 g, 119 mmol) portionwise so that the internal temperature remained below 10°C. After stirring for 4 hours at 0°C, the solution was poured over crushed ice and the resulting mixture was stirred. After 30 minutes, the organic phase was removed and the aqueous phase was extracted two times with dichloromethane. The combined organic phases were washed once with aqueous 1M hydrochloric acid solution, once with aqueous 1M sodium hydroxide solution, and once with brine. The organic phase was dried over sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was recrystallized from ethanol to afford (5-bromo-2-fluoro-phenyl)-(4-methoxy-phenyl)-methanone (22.5 g, 64%) as a white solid.
  • To a well stirred solution of (5-bromo-2-fluoro-phenyl)-(4-methoxy-phenyl)-methanone (22.5 g, 72.80 mmol) and triethylsilane (27.9 mL, 20.3 g, 175.0 mmol) in dichloromethane (20 mL) and acetonitrile (60 mL) at 0°C was added boron trifluoride etherate (32.0 mL, 36.2 g, 255.0 mmol) dropwise. Boron trifluoride etherate was added at a rate so that the internal temperature did not exceed 20°C. The reaction solution was warmed to room temperature and stirred overnight. After a total of 18 hours, a solution of potassium hydroxide (5.0 g) in water (15.0 mL) was added and the resulting mixture was stirred for 2 hours. The organic phase was separated and the aqueous phase was extracted two times with diethyl ether. The combined organic phases were washed once with aqueous 1M sodium hydroxide solution and once with brine. The organic phase was dried over sodium sulfate, filtered, and concentrated under reduced pressure. Upon addition of ethanol to the resulting residue a white solid formed. The solid was collected and dried under high vacuum to afford 4-bromo-1-fluoro-2-(4-methoxy-benzyl)-benzene (20.1 g, 93% yield) as a white solid.
  • 1H NMR (400 MHz, chloroform-d) delta ppm 3.79 (s, 3 H), 3.89 (s, 2 H), 6.85 (d, J=8.6 Hz, 2 H), 6.91 (t, 1 H), 7.12 (d, J=8.8 Hz, 2 H), 7.21 - 7.31 (m, 2 H).
  • Preparation of starting material 4-bromo-2-(4-ethoxy-benzyl)-benzonitrile:
  • A solution of ethyl (4-ethoxy-phenyl) acetate (2.68 g, 12.87 mmol), 4-bromo-2-fluoro-benzonitrile (2.74 g, 13.70 mmol) in N-methylpyrrolidone (4 mL) was slowly added to a suspension of potassium tert-butoxide (3.14 g, 27.98 mmol) in N-methylpyrrolidone (13 mL) at 0°C. Upon addition, the solution became dark red. The dark-red mixture was stirred at 0°C for 30 minutes and then at room temperature for 1 hour. Methanol (10 mL) and aqueous 1 M sodium hydroxide solution (13.7 mL) were added and the mixture was stirred overnight at room temperature. pH was adjusted to ~ 4 with hydrochloric acid (1 M aqueous solution) and the mixture was extracted with ethyl acetate (50 mL x 4). The combined organic layers were washed with brine, dried over sodium sulfate and evaporated to dryness. N,N-dimethylformamide (5 mL) and potassium carbonate (7 g) were added, the mixture was heated to 100°C for 1 hour and cooled to room temperature. Water was added and the mixture was extracted with ethyl acetate (60 mL x 3). The combined organic layers were washed with brine, dried over sodium sulfate and evaporated to dryness. The crude was purified by flash chromatography over silica gel (eluting with a gradient of 0 to 14% ethyl acetate in heptane), to afford 2.26 g of crude product (containing the desired product and another product). The crude product was precipitated with methanol, affording 4-bromo-2-(4-ethoxy-benzyl)-benzonitrile (1.2 g, containing 5 % of another compound with NMR peaks at 4.15 ppm quartet, and 1.5 ppm triplet).
  • 1H NMR (400 MHz, chloroform-d) delta 7.48-7.38 (m, 3 H), 7.13 (d, J = 8.4 Hz, 2H), 6.85 (d, J = 8.4 Hz, 2H), 4.08 (s, 2H), 4.03 (q, J = 7.2 Hz, 2H), 1.41 (t, J = 7.2 Hz, 3H).
  • Preparation of starting material 4-bromo-2-(4-ethoxy-benzyl)-1-fluoro-benzene
  • To a solution of 4-bromo-1-fluoro-2-(4-methoxy-benzyl)-benzene (4.2 g, 14.2 mmol) in dichloromethane (20 mL) at 0°C was slowly added a 1M solution of boron tribromide in dichloromethane (15.7 mL, 16.0 mmol) dropwise over 10 minutes. Once the addition of boron tribromide was complete the reaction mixture was gradually warmed to room temperature. After 4 hours, the reaction mixture was cooled to 0°C and quenched by slow addition of 1 N aqueous hydrochloric acid solution (20 mL). The reaction mixture was stirred for 30 minutes and extracted twice with dichloromethane. The combined organic layers were dried over magnesium sulfate, filtered, and concentrated under reduced pressure to afford a light pink solid (3.83 g, 96%). The crude product 4-(5-bromo-2-fluorobenzyl)-phenol was used in the next step without further purification.
    1H NMR (400 MHz, chloroform-d) delta ppm 3.88 (s, 2 H), 4.76 (br. s., 1 H), 6.77 (d, J=8.2 Hz, 2 H), 6.91 (t, J=9.1 Hz, 1 H), 7.07 (d, J=8.6 Hz, 2 H), 7.23 (dd, J=6.8, 2.3 Hz, 1 H), 7.26 - 7.31 (m, 1 H).
  • To a solution of 4-(5-bromo-2-fluoro-benzyl)-phenol (6.0 g, 21.0 mmol) in anhydrous N,N-dimethylformamide (20 mL) cooled at 0°C was added sodium hydride (60% dispersion in mineral oil, 1.02 g, 25.6 mmol). After stirring at 0°C for 45 minutes, iodoethane (2.08 mL, 25.6 mmol) was added dropwise and the resulting mixture was allowed to warm up to room temperature. After 18 hours, the reaction mixture was quenched with water and extracted twice with ethyl acetate. The combined organic layers were washed twice with water and once with brine, dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The crude residue was purified by flash chromatography over silica gel eluting with a gradient of 0 to 10% ethyl acetate in heptane to afford 4.6 g (58% yield) of the desired product as a yellow oil.
    1H NMR (400 MHz, chloroform-d) delta ppm 1.40 (t, J=7.0 Hz, 3 H), 3.89 (s, 2 H), 4.01 (q, J=6.9 Hz, 2 H), 6.83 (d, J=8.4 Hz, 2 H), 6.91 (t, J=9.0 Hz, 1 H), 7.10 (d, J=8.8 Hz, 2 H), 7.20 - 7.30 (m, 2 H).
  • Preparation of toluene-4-sulfonic acid tetrahydro-furan-3-yl ester
  • To a solution of 3-hydroxy tetrahydrofuran (2.5 g, 28.0 mmol) in anhydrous pyridine (60 mL) at room temperature was added 4-toluene sulfonyl chloride (6.49 g, 34.0 mmol). After stirring the reaction mixture for 18 hours at room temperature, the reaction mixture was concentrated under reduced pressure. The resulting residue was purified by flash chromatography over silica gel eluting with a gradient of 0 to 30% ethyl acetate in heptane to afford 3.5 g (51% yield) of the desired product as a colorless oil.
    1H NMR (400 MHz, chloroform-d) delta ppm 2.05 - 2.12 (m, 2 H), 2.45 (s, 3 H), 3.77 - 3.92 (m, 4 H), 5.09 - 5.14 (m, 1 H), 7.35 (d, J=8.00 Hz, 2 H), 7.79 (d, 2 H).
  • Preparation of toluene-4-sulfonic acid oxetan-3-yl ester
  • To a solution of oxetan-3-ol (1.0 g, 13.0 mmol) in anhydrous pyridine (25 mL) at room temperature was added 4-toluene sulfonyl chloride (3.09 g, 16.2 mmol). After stirring the reaction mixture for 18 hours at room temperature, the reaction mixture was concentrated under reduced pressure. The resulting residue was purified by flash chromatography over silica gel eluting with a gradient of 0 to 30% ethyl acetate in heptane to afford 1.9 g (62% yield) of the desired product as a white solid.
    1H NMR (400 MHz, chloroform-d) delta ppm 2.46 (s, 3 H), 4.63 - 4.75 (m, 4 H), 5.26 - 5.34 (m, 1 H), 7.36 (d, J=8.00 Hz, 2 H), 7.78 (d, J=8.40 Hz, 2 H).
  • Preparation of starting material 3-[4-(5-bromo-2-fluoro-benzyl)-phenoxy]-tetrahydro-furan
  • To a solution of 4-(5-bromo-2-fluoro-benzyl)-phenol (1.5 g, 5.3 mmol) and cesium carbonate (2.61 g, 8.0 mmol) in N,N-dimethylformamide (15.0 mL) at room temperature was added a solution of toluene-4-sulfonic acid tetrahydro-furan-3-yl ester (1.94 g, 8.0 mmol) in N,N-dimethyformamide (10.0 mL). The reaction mixture was then stirred overnight at 50°C. After a total of 18 hours, the reaction mixture was cooled to room temperature, diluted with brine and extracted 3 times with ethyl acetate. The combined organic layers were washed twice with water and once with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The resulting crude residue was purified by flash chromatography over silica gel eluting with a gradient of 0 to 30% ethyl acetate in heptane to afford 1.66 g (89% yield) of the desired product as a colorless oil.
    1H NMR (400 MHz, chloroform-d) delta ppm 2.09 - 2.24 (m, 2 H), 3.86 - 4.01 (m, 6 H), 4.86 - 4.91 (m, 1 H), 6.80 (d, J=8.6 Hz, 2 H), 6.91 (t, J=9 Hz, 1 H), 7.10 (d, J=8.6 Hz, 2 H), 7.23 (dd, J=6.8, 2.5 Hz, 1 H), 7.26 - 7.31 (m, 1 H).
  • Preparation of starting material 3-[4-(5-bromo-2-fluoro-benzyl)-phenoxy]-oxetane
  • To a solution of 4-(5-bromo-2-fluoro-benzyl)-phenol (1.1 g, 3.9 mmol) and cesium carbonate (1.91 g, 5.87 mmol) in N,N-dimethylformamide (15.0 mL) at room temperature was added a solution of toluene-4-sulfonic acid oxetan-3-yl ester (1.34 g, 8.0 mmol) in N,N-dimethylformamide (10.0 mL). The reaction mixture was then stirred overnight at 65°C. After a total of 18 hours, the reaction mixture was cooled to room temperature, diluted with brine and extracted 3 times with ethyl acetate. The combined organic layers were washed twice with water and once with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude residue was purified by flash chromatography over silica gel eluting with a gradient of 0 to 30% ethyl acetate in heptane to afford 0.948 g (72% yield) of the desired product as a white solid.
    1H NMR (400 MHz, chloroform-d) delta ppm 3.88 (s, 2 H), 4.76 (dd, J=7.22, 5.3 Hz, 2 H), 4.95 (t, J=6.6 Hz, 2 H), 5.14 - 5.21 (m, 1 H), 6.63 (d, J=8.4 Hz, 2 H), 6.92 (dd, 1 H), 7.10 (d, J=8.6 Hz, 2 H), 7.23 (dd, J=6.6, 2.15 Hz, 1 H), 7.26 - 7.31 (m, 1 H).
  • Preparation of 3-(4-(5-bromo-2-chlorobenzyl)phenoxy)oxetane:
  • 4-bromo-1-chloro-2-(4-methoxybenzyl)-benzene (10 g, 32 mmol) was dissolved in dichloromethane (32 mL) and cooled to 0°C under nitrogen. A 1.0 M boron tribromide solution in dichloromethane (35.3 mL, 34.3 mmol) was added dropwise over 10 minutes.
  • Following the addition, the ice bath was removed and the solution was stirred at room temperature for 1 hour. The reaction mixture was cooled to 0°C and quenched by addition of 1N aqueous hydrochloric acid solution (45 mL). The mixture was stirred for 30 minutes,
  • transferred to a separatory funnel, the organic layer was collected, and the aqueous layer was extracted with dichloromethane (45 mL). The combined organic extracts were dried over magnesium sulfate, filtered, and concentrated in vacuo to afford 4-(5-bromo-2-chlorobenzyl)phenol (9.5 g, 99% yield) as a white solid.
  • To a solution of crude 4-(5-bromo-2-chlorobenzyl)phenol (3.0 g, 10 mmol) and cesium carbonate (4.9 g, 15 mmol) in N,N-dimethylformamide (77.5 mL) at room temperature was added a solution of toluene-4-sulfonic acid oxetan-3-yl ester (3.5 g, 15 mmol) in N,N-dimethylformamide (8 mL). The mixture was heated to 65°C for 22 hours whereupon an additional aliquot of cesium carbonate (3.3 g, 10 mmol) was added. The reaction mixture was stirred for a further 12 hours at 120°C, cooled to room temperature whereupon water and ethyl acetate were added and the mixture was carefully acidified with 1N aqueous hydrochloric acid solution. The organic layer was separated, washed with brine (3 times), and concentrated in vacuo. Purification via Biotage MPLC (silica gel, eluting with a gradient of 0 to 25% ethyl acetate in heptane) afforded 3-(4-(5-bromo-2-chlorobenzyl)phenoxy)oxetane (2.5 g, 70% yield) as a white solid.
    1H NMR (400 MHz, dichloromethane-d2) delta ppm 7.34 - 7.28 (m, 2 H), 7.26 (d, J=8.4 Hz, 1 H), 7.14 - 7.09 (m, 2 H), 6.69 - 6.35 (m, 2 H), 5.22 - 5.16 (m, 1 H), 4.96 - 4.91 (m, 2H), 4.72 - 4.68 (m, 2H), 4.01 (s, 2H).
  • Preparation of 4-bromo-2-(4-chloro-benzyl)-1-fluoro-benzene
  • A solution of 5-bromo-2-fluorobenzaldehyde (10.2 g, 50 mmol) in anhydrous tetrahydrofuran (200 mL) was cooled to -78°C. A solution of 4-chlorophenyl-magnesium bromide (1M in diethyl ether, 60 mL, 60 mmol) was added via syringe over 8 minutes. Stirring was continued at low temperature for 5 minutes and the reaction was warmed up to room temperature and stirred for 1 hour at this temperature. The solution was cooled in an ice-water bath and quenched by addition of saturated aqueous ammonium chloride solution (40 mL). The organic phase was decanted and the aqueous residue was concentrated under reduced pressure to remove any remaining organic solvent. The aqueous phase was extracted with ethyl acetate (200 mL × 2) and the extracts were combined with the decanted tetrahydrofuran solution. This solution was washed with brine (25 mL) and was dried (sodium sulfate), filtered and concentrated under reduced pressure, giving crude (5-bromo-2-fluorophenyl)-(4-chlorophenyl)-methanol (15.2 g, 96% yield) as a yellow solid.
  • To a solution of the above (5-bromo-2-fluorophenyl)-(4-chlorophenyl)-methanol (15.0 g, 48 mmol) and triethylsilane (18.5 mL, 116 mmol) in dichloromethane (40 mL) and acetonitrile (20 mL) at 0°C under nitrogen, was slowly added boron trifluoride diethyl etherate (22.7 mL, 181 mmol). The resulting solution was stirred for 18 hours, while slowly warming to room temperature. The reaction was cooled in an ice-water bath, quenched by slow addition of 7 M aqueous potassium hydroxide solution (30 mL) and extracted with methyl tert-butyl ether (200 mL × 2). The combined organic solution was washed with water (25 mL × 2), brine (25 mL × 2), dried (sodium sulfate), filtered and concentrated under reduced pressure. Purification by flash column chromatography over silica gel eluting with a gradient of ethyl acetate in heptane gave 2-(4-chlorobenzyl)-4-bromo-1-fluorobenzene (5.0 g, 35% yield) as a colorless oil. 1H NMR (400 MHz, chloroform-d) delta ppm 7.33-7.22 (m, 4H), 7.13 (d, J = 8.4 Hz, 2H), 6.93 (dd, J = 9.2, 9.2 Hz, 1H), 3.92 (s, 2H).
  • Preparation of Intermediates Preparation of Intermediate ((2R,3R,45,5R)-6-allyloxy-3,4,5-tris-benzyloxy-tetrahydropyran-2-yl)-methanol (I-1a):
  • Figure imgb0009
  • A suspension of D-glucose (1.2 kg, 6.6 mol), trifluoromethane sulfonic acid (12 mL) and allyl alcohol (5 L) was heated at 80°C for 3 days. The mixture was cooled down to room temperature, the volatiles were removed in vacuo and the residue dissolved in N,N-dimethylformamide (8 L). This was split into four equal reactions and to each was added trityl chloride (463 g, 1.67 mol) and triethylamine (231 mL, 1.67 mol). A slight exotherm was observed while adding the triethylamine. The reaction mixture was stirred for 2 days at 30°C and then each reaction split in half, giving eight equal reactions. To each of these reactions was added benzyl chloride (300 mL, 2.60 mol), followed by portionwise addition of sodium hydride (102.5 g, 2.60 mol) maintaining the reaction temperature between 40 to 50°C. After complete addition, the reaction mixtures were stirred at room temperature for 20 hours. Each reaction was then poured onto ice/water (2 L) and extracted with ethyl acetate (2.5 L). The organic phases of each were washed with saturated brine/water (1:1, 2 x 2 L), combined and dried over magnesium sulfate (product Rf 0.85 in 3:1 hexanes/ethyl acetate) After filtration and evaporation the residue was dissolved in a mixture of dichloromethane (16 L) and methanol (4 L). The mixture was split into 5 equal portions and to each was added sulfuric acid (32 mL). The reactions were stirred for 3 hours, washed with brine/2M aqueous sodium hydroxide solution (1:1, 2 x 2 L), combined and dried over magnesium sulfate. After filtration and concentration in vacuo, the residue was further purified on silica gel eluting with 30% ethyl acetate in toluene to give intermediate compound (I-1a) as a mixture of anomers (1.77 kg, 54 % yield from D-glucose). Rf 0.15 in 3:1 hexanes/ethyl acetate.
  • Preparation of Intermediate (13S,4S,5R)-6-allyloxy-3,4,5-tris-benzyloxy-2-hydroxymethyl-tetrahydro-pyran-2-yl)-methanol (I-1b):
  • Figure imgb0010
  • A solution of dimethylsulfoxide (87 mL, 1.22 mol) in dichloromethane (160 mL) was added dropwise to a solution of oxalyl chloride (64.7 mL, 0.76 mol) in dichloromethane (2.5 L) at -78°C. After complete addition a solution of intermediate (I-1a) (287 g, 0.59 mol) in dichloromethane (500 mL) was added dropwise at -78°C. After complete addition the reaction mixture was stirred for 30 minutes and triethylamine (417 mL, 2.9 mol) was added dropwise. After complete addition the reaction mixture was allowed to self warm to room temperature. The reaction was then washed with 1 M aqueous hydrochloric acid solution (2 L) and water (2 L), and then dried over magnesium sulfate. This reaction procedure was repeated on six equivalent reactions and after drying they were combined and evaporated to give the aldehyde as a yellow oil (1.71 kg). This oil was dissolved in isopropanol (2.57 L) and split into seven equal reactions. To each of these was added a 37% aqueous formaldehyde solution (0.79 L, 10 mol), followed by the dropwise addition of a solution of sodium hydroxide (32 g, 0.8 mol) in water (130 mL). After complete addition the reaction mixture was stirred at room temperature for 2 days. The reaction mixture was diluted with brine (2 L) and extracted with ethyl acetate (2 L). The organic phase was further washed with saturated aqueous sodium bicarbonate solution (2 L), brine (2 L) and then dried over magnesium sulfate. The organic phases from the seven reactions were combined, evaporated and the residue purified on silica gel (eluting with 4 to 1 up to 1 to 1 hexanes in ethyl acetate) to give intermediate compound (I-1b) as a mixture of anomers (980 g, 53% yield over the two steps). Rf 0.57 and 0.60 in 1:1 hexanes/ethyl acetate.
  • (3S,4S,5R)-6-allyloxy-3,4,5-tris-benzyloxy-2,2-bis-(4-methoxy-benzyloxymethyl)-tetrahydropyran (I-1c):
  • Figure imgb0011
  • The starting diol [((3S,4S,5R)-6-allyloxy-3,4,5-tris-benzyloxy-2-hydroxymethyl-tetrahydropyran-2-yl)-methanol (I-1b: 10 g, 19.208 mmol) was dissolved in N,N-dimethylformamide (70 mL) and cooled to 0°C. Sodium hydride (60% dispersion in mineral oil, 1.69 g, 42.3 mmol) was added and the reaction was allowed to stir at 0°C for 1 hour before the addition of 1-bromomethyl-4-methoxy-benzene (5.96 mL, 40.3 mmol). The reaction was then heated to 60°C overnight. The mixture was cooled down to room temperature and the reaction was quenched with water and extracted with ethyl acetate (2 times). The combined organic layers were washed with water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The reaction was then chromatographed over silica gel (eluting with a gradient of 0 to 80% ethyl acetate in heptane) yielding 7.55 g (52% yield) of product (I-1c). MS 778.8 (M + NH4 +; positive mode).
  • (3R,4S,5S)-3,4,5-tris-benzyloxy-6,6-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-Dyran-2-ol (I-1d):
  • Figure imgb0012
  • To a solution of starting material ((3S,4S,5R)-6-allyloxy-3,4,5-tris-benzyloxy-2,2-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-pyran, (I-1c: 7.55 g, 9.92 mmol) in methanol (60 mL) and dichloromethane (20 mL) at room temperature was added in palladium (II) chloride (528 mg, 2.98 mmol) and the resulting mixture was stirred at this temperature for 4 hours. TLC indicated the clean formation of a more polar product. The reaction was filtered through Celite® and concentrated under reduced pressure. The crude material was chromatographed over silica gel eluting with a gradient of 0 to 80% ethyl acetate in heptane yielding 5.6 g (78% yield) of product (L-1d). MS 738.8 (M + NH4 +; positive mode).
  • (3R,45,5S)-3,4,5-tris-benzyloxy-6,6-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-pyran-2-one (I-1e):
  • Figure imgb0013
  • To a solution of oxalyl dichloride (1.9 mL, 23 mmol) in dichloromethane (65 mL) at -78°C was added a solution of dimethyl sulfoxide (3.3 mL, 47 mmol) in dichloromethane (5 mL) and the resulting solution was stirred at this temperature for 30 minutes. A solution of starting material ((3R,4S,5S)-3,4,5-tris-benzyloxy-6,6-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-pyran-2-ol, (I-1d, 5.6 g, 7.7 mmol) in dichloromethane (15.0 mL) was then added dropwise and the resulting mixture was stirred for 30 minutes allowing the temperature to rise to -60°C. Triethylamine (9.7 mL, 69.5 mmol) was added dropwise and the mixture allowed to warm up to 0°C over 1 hour. The reaction was quenched by addition of saturated aqueous ammonium chloride solution and the organic phase was dried over magnesium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by flash chromatography over silica gel eluting with a gradient of 0 to 60% ethyl acetate in heptane to produce the product (I-1e) (4 g, 72% yield).
    1H NMR (400 MHz, chloroform-d) delta ppm 3.24 (d, J=10 Hz, 1 H), 3.40 - 3.47 (m, 2 H), 3.74 (s, 3 H), 3.77 (s, 3 H), 3.86 (d, J=10 Hz, 1 H), 4.07 (d, J=8.6 Hz, 1 H), 4.15 (d, J=9.6 Hz, 1 H), 4.35 - 4.55 (m, 6 H), 4.65 - 4.72 (m, 2 H), 4.82 (d, J=11 Hz,1 H), 4.87 (d, J=11.2 Hz, 1 H), 5.10 (d, J=11.1 Hz, 1 H), 6.74 - 6.79 (m, 2 H), 6.81 - 6.85 (m, 2 H), 7.11 (dd, J=7.0, 2.5 Hz, 2 H), 7.17 - 7.41 (m, 17 H).
  • (2R,3S,4S)-2,3,4-tris-benzyloxy-5-hydroxy-6-(4-methoxy-benzyloxy)-5-(4-methoxy-benzyloxymethyl)-hexanoic acid methoxy-methyl-amide (I-1g) and/or (3R,4S,5S)-3,4,5-tris-benzyloxy-6,6-bis-(4-methoxy-benzyloxymethy/)-2-(methoxy-methyl-amino)-tetrahydropyran-2-ol (I-1f):
  • Figure imgb0014
  • To a solution of lactone ((3R,4S,5S)-3,4,5-tris-benzyloxy-6,6-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-pyran-2-one (I-1e: 10.4 g, 14.5 mmol) and N,O-dimethylhydroxylamine hydrochloride (1.77 g, 29.0 mmol) in dichloromethane (100 mL) at 0°C was added dropwise a 2.0 M solution of trimethyl aluminum in hexanes (14.5 mL, 29.0 mmol) and the resulting solution was stirred at room temperature for 16 hours. The reaction mixture was cooled to 0°C and quenched by slow addition of aqueous 1N hydrochloric acid solution. The resulting mixture was allowed to stir for 1 hour. The organic phase was separated and washed with aqueous 1N hydrochloric acid solution, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude material was purified by medium pressure chromatography (gradient of 5 to 40% ethyl acetate in heptane) yielding 6.5 g (58%) of product.
    1H NMR (400 MHz, chloroform-d) delta ppm 2.62 (br. s, 1 H), 2.94 (br. s., 3 H), 3.23 (br. s., 3 H), 3.42 (d, J=9.4 Hz, 1 H), 3.50 - 3.60 (m, 3 H), 3.75 (s, 3 H), 3.77 (s, 3 H), 4.03 (d, J=6.9 Hz, 1 H), 4.20 (dd, J=6.9, 3.3 Hz, 1 H), 4.31 - 4.44 (m, 5 H), 4.46 - 4.51 (m, 2H), 4.53 (d, J=12 Hz, 1 H), 4.66 (d, J=12 Hz, 1 H), 4.80 (br. d, J=11.5 Hz, 1 H), 4.87 (d, J=11.4 Hz, 1 H), 6.77 - 6.83 (m, 4 H), 7.15 - 7.35 (m, 19 H). ([M+H+] 780.8, positive mode; [M+HCO2] 824.7, negative mode). HRMS calculated for C46H54NO10 (M+H+) 780.3742, found 780.3708.
  • (2R,3S,4S)-2,3,4,6-tetrakis-benzyloxy-5-benzyloxymethyl-5-hydroxy-hexanoic acid methoxy-methyl-amide (I-6g) and/or (3R,4S,5S)-3,4,5-tris-benzyloxy-6,6-bis-benzyloxymethyl-2-(methoxy-methyl-amino)-tetrahydro-pyran-2-ol (I-6f):
  • Figure imgb0015
  • This compound was prepared starting from [((3S,4S,5R)-6-allyloxy-3,4,5-tris-benzyloxy-2-hydroxymethyl-tetrahydro-pyran-2-yl)-methanol (I-1b) using a similar procedure as the one described for the synthesis of (2R,3S,4S)-2,3,4-tris-benzyloxy-5-hydroxy-6-(4-methoxy-benzyloxy)-5-(4-methoxy-benzyloxymethyl)-hexanoic acid methoxy-methyl-amide (I-1g) and/or (3R,4S,5S)-3,4,5-tris-benzyloxy-6,6-bis-(4-methoxy-benzyloxymethyl)-2-(methoxy-methyl-amino)-tetrahydro-pyran-2-ol (I-1f) except that the alkylating agent used in the experimental part describing the conversion from (I-1 b) to (I-1c) was benzyl bromide instead of para-methoxybenzyl bromide.
    1H NMR (400 MHz, chloroform-d) delta ppm 2.66 (br. s, 1 H), 2.94 (br. s., 3 H), 3.23 (br. s., 3 H), 3.48 (d, J=9.4 Hz, 1 H), 3.55 - 3.66 (m, 3 H), 4.05 (d, J=6.9 Hz, 1 H), 4.21 (dd, J=6.9, 3.3 Hz, 1 H), 4.36 (d, 1 H, J = 11.7 Hz), 4.41 - 4.58 (m, 7 H), 4.68 (d, J=11.9 Hz, 1 H), 4.81 (br. d, J=11.5 Hz, 1 H), 4.89 (d, J=11.5 Hz, 1 H), 7.15 - 7.35 (m, 25 H). MS [M+H+] 720.7, positive mode; [M+HCO2] 764.7, negative mode).
  • (4S,5S)-3,4,5-tris-benzyloxy-2-[3-(4-methoxy-benzyl)-4-methyl-phenyl]-6,6-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-pyran-2-ol (I-1i) :
  • Figure imgb0016
  • n-Butyl lithium (0.97 mL, 2.5 M/hexanes, 3.15 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution (placed in a pre dried Biotage microwave vial 10-20 mL sealed with its cap and placed under a positive stream of nitrogen gas) of 4-bromo-2-(4-methoxy-benzyl)-1-methyl-benzene (690 mg, 3 equivalents) in anhydrous tetrahydrofuran (2.7 mL) at -78°C and the resulting solution was stirred at this temperature for an additional hour. A solution of (2R,3S,4S)-2,3,4-tris-benzyloxy-5-hydroxy-6-(4-methoxy-benzyloxy)-5-(4-methoxy-benzyloxymethyl)-hexanoic acid methoxy-methyl-amide (I-1g) (608 mg) in anhydrous tetrahydrofuran (1.35 mL) was then added dropwise over 1.5 hours using a syringe pump and the resulting mixture was stirred at - 78°C for 1 hour before being allowed to warm to -20°C over 14 hours (put in a deep Dewar covered with aluminum foil to maintain cold temperature; size of Dewar: external diameter 10 cm, internal diameter 8 cm, height 9 cm). Diethyl ether was added and the reaction was quenched by dropwise addition of 1 M aqueous hydrochloric acid solution. The resulting biphasic mixture was stirred at room temperature for 15 minutes. The organic phase was separated, washed with brine, dried over magnesium sulfate, filtered and concentrated. Chromatography over silica gel using a gradient of 20 to 50% ethyl acetate in heptane gave the product as a mixture of isomers (440 mg, 61% yield).
    HRMS calculated for C59H62O10Na (M+Na+) 953.4235, found 953.4236.
  • {(2S,3S)-2,3, 4-tris-benzyloxy-5-[3-(4-methoxy-benzyl)-4-methyl-phenyl]-6,8-dioxa-bicyclo[3.2.1]oct-1-yl}-methanol (I-1k):
  • Figure imgb0017
  • To a solution of intermediate I-1i (150 mg) in dichloromethane (3 mL) was added anisole (90 microL, 5 equivalents) followed by 3 mL of a solution of 20% trifluoroacetic acid in dichloromethane and the resulting mixture was stirred at room temperature for about 1 hour. The mixture was concentrated and the crude was chromatographed over silica gel (using a gradient of 10 to 30% ethyl acetate in heptane) to afford the desired product as a mixture of isomers (66 mg, 61% yield). MS (LCMS) 673.9 (M+H+; positive mode).
  • (4S, 5S)-3,4,5-tris-benzyloxy-2-(3-(4-ethoxy-benzyl)-4-methyl-phenyl]-6,6-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-pyran-2-ol (I-2i):
  • Figure imgb0018
  • n-Butyl lithium (0.312 mL, 2.5 M/hexanes, 3.05 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution (placed in a pre dried Biotage microwave vial 10-20 mL sealed with its cap and placed under a positive stream of nitrogen gas) of 4-bromo-2-(4-ethoxy-benzyl)-1-methyl-benzene (238 mg, 3.05 equivalents) in anhydrous tetrahydrofuran (0.9 mL) at -78°C and the resulting solution was stirred at this temperature for an additional hour. A solution of (2R,3S,4S)-2,3,4-tris-benzyloxy-5-hydroxy-6-(4-methoxy-benzyloxy)-5-(4-methoxy-benzyloxymethyl)-hexanoic acid methoxy-methyl-amide (I-1g) (200 mg) in anhydrous tetrahydrofuran (0.6 mL) was then added dropwise over 1.5 hours using a syringe pump and the resulting mixture was stirred at - 78°C for 1 hour before being allowed to warm to room temperature over 16 hours (put in a deep Dewar covered with aluminum foil to maintain cold temperature; size of Dewar: external diameter 10 cm, internal diameter 8 cm, height 9 cm). Diethyl ether was added and the reaction was quenched by dropwise addition of aqueous 1 M hydrochloric acid solution. The resulting biphasic mixture was stirred at room temperature for 15 minutes. The organic phase was separated, washed with brine, dried over magnesium sulfate, filtered and concentrated. The crude material was chromatographed using the Biotage automated chromatography unit (two stacked 10g silica gel columns; eluting with a gradient of 0 to 60% ethyl acetate in heptane) to give the product as a mixture of isomers (136 mg, 56% yield). MS (LCMS) 968 (M+Na+; positive mode).
  • {(2S,3S)-2,3,4-tris-benzyloxy-5-[3-(4-ethoxy-benzyl)-4-methyl-phenyl]-6,8-dioxa-bicyclo[3.2.1]oct-1-yl}-methanol (I-2k):
  • Figure imgb0019
  • To a solution of the intermediate I-2i (136 mg, 0.145 mmol) in dichloromethane (4 mL) was added anisole (310 microL, ~5 equivalents) followed by 4 mL of a solution of 20% trifluoroacetic acid in dichloromethane and the resulting mixture was stirred at room temperature for 1.5 hours. The mixture was concentrated and the crude was chromatographed using the ISCO combiflash® companion® automated chromatography unit (4g silica gel column) and eluting with a gradient of 0 to 70% ethyl acetate in heptane to afford the desired product as a mixture of isomers (85 mg, 85% yield). MS (LCMS) 687.7 (M+H+; positive mode).
  • (4S,5S)-3,4,5-tris-benzyloxy-2-[4-chloro-3-(4-methoxy-benzyl)-phenyl]-6,6-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-pyran-2-ol (I-3i):
  • Figure imgb0020
  • n-Butyl lithium (0.97 mL, 2.5 M/hexanes, 3.15 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution (placed in a pre dried Biotage microwave vial 10-20 mL sealed with its cap and placed under a positive stream of nitrogen gas) of 4-bromo-1-chloro-2-(4-methoxy-benzyl)-benzene (725 mg, 2.95 equivalents) in anhydrous tetrahydrofuran (2.7 mL) at -78°C and the resulting solution was stirred at this temperature for an additional hour. A solution of (2R,3S,4S)-2,3,4-tris-benzyloxy-5-hydroxy-6-(4-methoxy-benzyloxy)-5-(4-methoxy-benzyloxymethyl)-hexanoic acid methoxy-methyl-amide (I-1g) (616 mg) in anhydrous tetrahydrofuran (1.35 mL) was then added dropwise over 1.5 hours using a syringe pump and the resulting mixture was stirred at - 78°C for 1 hour before being allowed to warm to -20°C over 14 hours (put in a deep Dewar covered with aluminum foil to maintain cold temperature; size of Dewar: external diameter 10 cm, internal diameter 8 cm, height 9 cm). Diethyl ether was added and the reaction was quenched by dropwise addition of aqueous 1 M hydrochloric acid solution. The resulting biphasic mixture was stirred at room temperature for 15 minutes. The organic phase was separated, washed with brine, dried over magnesium sulfate, filtered and concentrated. Chromatography over silica gel using a gradient of 10 to 40% ethyl acetate in heptane gave the product as a mixture of isomers (530 mg, 71 % yield).
  • {(2S,3S)-2,3,4-tris-benzyloxy-5-(4-chloro-3-(4-methoxy-benzyl)-phenyl]-6,8-dioxa-bicyvclo[3.2.1]oct-1-yl}-methanol (I-3k):
  • Figure imgb0021
  • To a solution of the intermediate I-3i (530 mg) in dichloromethane (11 mL) was added anisole (300 microL, 5 equivalents) followed by 11 mL of a solution of 20% trifluoroacetic acid in dichloromethane and the resulting mixture was stirred at room temperature for 1 hour. The mixture was concentrated and the crude was chromatographed over silica gel using a gradient of 10 to 40% ethyl acetate in heptane to afford the product as a mixture of isomers (229 mg, 59% yield).
    MS (LCMS) 693.6 (M+H+; positive mode).
  • (4S,5S)-3,4,5-tris-benzyloxy-2-[4-chloro-3-(4-ethoxy-benzyl)-phenyl)-6,6-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-pyran-2-ol (I-4i):
  • Figure imgb0022
  • n-Butyl lithium (1.0 mL, 2.5 M/hexanes, 3.25 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution (placed in a pre dried Biotage microwave vial 10-20 mL sealed with its cap and placed under a positive stream of nitrogen gas) of 4-bromo-1-chloro-2-(4-ethoxy-benzyl)-benzene (815 mg, 3.25 equivalents) in anhydrous tetrahydrofuran (2.9 mL) at -78°C and the resulting solution was stirred at this temperature for an additional hour. A solution of (2R,3S,4S)-2,3,4-tris-benzyloxy-5-hydroxy-6-(4-methoxy-benzyloxy)-5-(4-methoxy-benzyloxymethyl)-hexanoic acid methoxy-methyl-amide (I-1g) (600 mg) in anhydrous tetrahydrofuran (1.45 mL) was then added dropwise over 1.3 hours using a syringe pump and the resulting mixture was stirred at - 78°C for 1 hour before being allowed to warm to -25°C over 14 hours (put in a deep Dewar covered with aluminum foil to maintain cold temperature; size of Dewar: external diameter 10 cm, internal diameter 8 cm, height 9 cm). Diethyl ether was added and the reaction was quenched by dropwise addition of aqueous 1 M hydrochloric acid solution. The resulting biphasic mixture was stirred at room temperature for 15 minutes. The organic phase was separated, washed with brine, dried over magnesium sulfate, filtered and concentrated. Chromatography over silica gel using a gradient of 10 to 40% ethyl acetate in heptane gave the product as a mixture of isomers (280 mg, 38% yield).
    HRMS calculated for C59H61O10ClNa (M+Na+) 987.3845, found 987.3840.
  • {(2S,3S)-2,3,4-tris-benzyloxy-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-6,8-dioxa-bicyclo[3.2.1]oct-1-yl]-methanol (I-4k):
  • Figure imgb0023
  • To a solution of the intermediate I-4i (1.46 g) in dichloromethane (31 mL) was added anisole (900 microL, ~5 equivalents) followed by 31 mL of a solution of 20% trifluoroacetic acid in dichloromethane and the resulting mixture was stirred at room temperature for 1 hour. The mixture was concentrated and the crude was chromatographed over silica gel using a gradient of 10 to 30% ethyl acetate in heptane to afford the product as a mixture of isomers (670 mg, 63% yield).
    HRMS calculated for C43H44O7Cl (M+H+) 707.2770, found 707.2765.
  • (4S,5S)-3,4,5-tris-benzyloxy-2-[4-fluoro-3-(4-methoxy-benzyl)-phenyl]-6,6-bis-(4methoxy-benzyloxymethyl)-tetrahydro-pyran-2-ol (I-5i):
  • Figure imgb0024
  • n-Butyl lithium (462 microL, 2.5 M/hexanes, 3.0 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution (placed in a pre dried Biotage microwave vial 10-20 mL sealed with its cap and placed under a positive stream of nitrogen gas) of 4-Bromo-1-fluoro-2-(4-methoxy-benzyl)-benzene (341 mg, 3 equivalents) in anhydrous tetrahydrofuran (1.4 mL) at -78 °C under nitrogen. The resulting solution was stirred at this temperature for 1 hour. Then a solution of (2R,3S,4S)-2,3,4-tris-benzyloxy-5-hydroxy-6-(4-methoxy-benzyloxy)-5-(4-methoxy-benzyloxymethyl)-hexanoic acid methoxy-methyl-amide (I-1g) (300 mg, 0.385 mmol) in anhydrous tetrahydrofuran (0.70 mL) was added dropwise very slowly (1 drop every 5 seconds) and the resulting mixture was stirred at -78°C for an additional hour before warming to 10 °C over 12 hours (put in a deep Dewar covered with aluminum foil to maintain cold temperature; size of Dewar: external diameter 10 cm, internal diameter 8 cm, height 9 cm). The reaction was diluted with diethyl ether and quenched by the dropwise addition of aqueous 1 N hydrochloric acid solution. The resulting biphasic mixture was stirred at room temperature for 15 minutes. The organic phase was separated, washed with brine, dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The crude residue was purified by flash chromatography over silica gel (eluting with a gradient of 10 to 40% ethyl acetate in heptane) to afford the product as a mixture of isomers (199 mg, 55 % yield).
  • {(2S,3S)-2,3,4-tris-benzyloxy-5-[4-fluoro-3-(4-methoxy-benzyl)-phenyl]-6,8-dioxa-bicyclo(3.2.1]oct-1-yl}-methanol (I-5k):
  • Figure imgb0025
  • To a solution of (4S,5S)-3,4,5-Tris-benzyloxy-2-[4-fluoro-3-(4-methoxy-benzyl)-phenyl]-6,6-bis-(4methoxy-benzyloxymethyl)-tetrahydro-pyran-2-ol (I-5i; 191 mg, 0.204 mmol) in dichloromethane (3.75 mL) was added anisole (0.178 mL, 1.63 mmol) followed by a 20% solution of trifluoroacetic acid in dichloromethane (3.75 mL) at room temperature under nitrogen. After stirring for 1 hour at room temperature, the reaction mixture was concentrated under reduced pressure. The crude residue was purified by flash chromatography over silica gel (eluting with a gradient of 10 to 30% ethyl acetate in heptane) to afford the product as a mixture of isomers (115 mg, 83% yield). MS (LCMS) 677.7 (M+H+; positive mode).
  • (4S,5S)-3,4,5-tris-benzyloxy-2-[3-(4-ethoxy-benzyl)-4-fluoro-phenyil-6,6-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-pyran-2-ol (I-10i)
  • Figure imgb0026
  • n-Butyl lithium (508 microL, 2.5 M/hexanes, 3.0 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution of 4-Bromo-2-(4-ethoxy-benzyl)-1-fluorobenzene (392.0 mg, 1.27 mmol) in anhydrous tetrahydrofuran (1.5 mL) at -78°C under nitrogen. The resulting solution was stirred at this temperature for 1 hour. Then a solution of (2R,3S,4S)-2,3,4-tris-benzyloxy-5-hydroxy-6-(4-methoxy-benzyloxy)-5-(4-methoxy-benzyloxymethyl)-hexanoic acid methoxy-methyl-amide I-1g (330.0 mg, 0.423 mmol) in anhydrous tetrahydrofuran (0.75 mL) was added dropwise very slowly (1 drop every 5 seconds) and the resulting mixture was stirred at -78°C for an additional hour before warming to 10°C over 12 hours (put in a deep Dewar covered with aluminum foil to maintain cold temperature). The reaction was diluted with diethyl ether and quenched by the dropwise addition of aqueous 1 N hydrochloric acid solution. The resulting biphasic mixture was stirred at room temperature for 15 minutes. The organic phase was separated, washed with brine, dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The crude residue was purified by flash chromatography over silica gel (eluting with a gradient of 10 to 40% ethyl acetate in heptane) to afford the product as a mixture of isomers (180 mg, 44% yield).
  • {(2S,3S)-2,3,4-tris-benzyloxy-5[3-(4-ethoxy-benzyl)-4-fluoro-phenyl]-6,8-dioxa-bicyclo[3.2.1)oct-1-yl}-methanol (I-10k)
  • Figure imgb0027
  • To a solution of intermediate I-10i (180.0 mg, 0.19 mmol) in dichloromethane (2.0 mL) was added anisole (0.175 mL, 1.60 mmol) followed by a 20% solution of trifluoroacetic acid in dichloromethane (2.0 mL) at room temperature under nitrogen. After stirring for 1 hour, the reaction mixture was concentrated under reduced pressure. The crude residue was purified by flash chromatography over silica gel (eluting with a gradient of 10 to 30% ethyl acetate in heptane) to afford the product as a mixture of isomers (85.0 mg, 64% yield).
  • (4S,5S)-3,4,5-tris-benzyloxy-2-{4-fluoro-3-[4-(tetrahydro-furan-3-yloxy)-benzyl]-phenyl}-6,6-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-pyran-2-ol (I-11i)
  • Figure imgb0028
  • n-Butyl lithium (1.0 mL, 2.5 M/hexanes, 3.0 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution of 3-[4-(5-bromo-2-fluoro-benzyl)-phenoxy]-tetrahydro-furan (878 mg, 2.50 mmol) in anhydrous tetrahydrofuran (3.0 mL) at -78°C and the resulting solution was stirred at this temperature for 1 hour. A solution of (2R,3S,4S)-2,3,4-tris-benzyloxy-5-hydroxy-6-(4-methoxy-benzyloxy)-5-(4-methoxy-benzyloxymethyl)-hexanoic acid methoxy-methyl-amide I=1g (650 mg, 0.833 mmol) in anhydrous tetrahydrofuran (1.5 mL) was then added dropwise very slowly (0.9 mL/hour) and the resulting mixture was stirred at -78°C for an additional hour before warming to 10°C over 12 hours (put in a deep Dewar covered with aluminum foil to maintain cold temperature). The reaction was diluted with diethyl ether and quenched by the dropwise addition of aqueous 1 N hydrochloric acid solution. The resulting biphasic mixture was stirred at room temperature for 15 minutes. The organic phase was separated, washed with brine, dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The crude residue was purified by flash chromatography over silica gel (eluting with a gradient of 10 to 40% ethyl acetate in heptane) to afford the product as a mixture of isomers (287 mg, 34% yield)
  • ((2S,3S)-2,3,4-tris-benzyloxy-5-{4-fluoro-3-[4-(tetrahydro-furan-3-yloxy)-benzyl]-phenyl}-6,8-dioxa-bicyclo[3.2.1]oct-1-yl)-methanol (I-11k)
  • Figure imgb0029
  • To a solution of (4S,5S)-3,4,5-Tris-benzyloxy-2-{4-fluoro-3-[4-(tetrahydro-furan-3-yloxy)-benzyl]-phenyl}-6,6-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-pyran-2-ol I-11i (275 mg, 0.28 mmol) in dichloromethane (2.0 mL) was added anisole (0.250 mL, 2.29 mmol) followed by a 20% solution of trifluoroacetic acid in dichloromethane (8.0 mL) at room temperature under nitrogen. After stirring for 1 hour, the reaction mixture was concentrated under reduced pressure. The crude residue was purified by flash chromatography over silica gel (eluting with a gradient of 10 to 30% ethyl acetate in heptane) to afford the product as a mixture of isomers (168 mg, 83% yield).
  • (4S,5S)-3,4,5-tris-benzyloxy-2-[3-(4-chloro-benzyl)-4-fluoro-phenyl]-6,6-bis-(4-methoxy-benzvloxymethyl)-tetrahydro-pyran-2-ol (I-12i):
  • Figure imgb0030
  • n-Butyl lithium (1.0 mL, 2.5 M/hexanes, 3.1 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution (placed in a pre dried Biotage microwave vial 10-20 mL sealed with its cap and placed under a positive stream of nitrogen gas) of 4-Bromo-2-(4-chloro-benzyl)-1-fluoro-benzene (702 mg, 2.9 equivalents) in anhydrous tetrahydrofuran (3.0 mL) at -78°C and the resulting solution was stirred at this temperature for 25 minutes. A solution of (2R,3S,4S)-2,3,4-tris-benzyloxy-5-hydroxy-6-(4-methoxy-benzyloxy)-5-(4-methoxy-benzyloxymethyl)-hexanoic acid methoxy-methyl-amide (I-1g) (621 mg) in anhydrous tetrahydrofuran (1.5 mL) was then added dropwise using a syringe pump (0.9 mL/hour) and the resulting mixture was stirred at low temperature for an additional 17 hours (placed in a deep Dewar covered with aluminum foil to maintain cold temperature; size of Dewar: external diameter 10 cm, internal diameter 8 cm, height 9 cm). The reaction was quenched by dropwise addition of aqueous 1 M hydrochloric acid solution (1.5 mL). The resulting biphasic mixture was stirred at room temperature for 30 minutes. The mixture was diluted with saturated aqueous ammonium chloride (15 mL) and was extracted with ethyl acetate (15 mL x 3). The combined organic solution was washed with brine (30 mL), dried over magnesium sulfate, filtered and concentrated. Chromatography over silica gel using a gradient of 10 to 40% ethyl acetate in heptane gave the product as a mixture of isomers (477 mg, 64% yield).
  • {(2S,3S)-2,3,4-tris-benzyloxy-5-[3-(4-chloro-benzyl)-4-fluoro-phenyl]-6,8-dioxa-bicyclo[3.2.1]oct-1-yl}-methanol (I-12k):
  • Figure imgb0031
  • To a solution of the intermediate I-12i (243 mg) in dichloromethane (9 mL) was added anisole (0.15 mL, 5.3 equivalents) followed by trifluoroacetic acid (1.0 mL, 50 equivalents) and the resulting mixture was stirred at room temperature for 2 hours. The mixture was concentrated and the crude was chromatographed over silica gel using a gradient of 10 to 30% ethyl acetate in heptane to afford the product as a mixture of isomers (102 mg, 58% yield).
  • (4S,5S)-3,4,5-tris-benzyloxy-2-{4-fluoro-3-[4-(oxetan-3-yloxy)-benzyl]-phenyl]-6,6-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-pyran-2-ol (I-13i)
  • Figure imgb0032
  • n-Butyl lithium (1.12 mL, 2.5 M/hexanes, 3.0 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution of 3-[4-(5-Bromo-2-fluoro-benzyl)-phenoxy]-oxetane (942.0 mg, 2.79 mmol) in anhydrous tetrahydrofuran (3.0 mL) at -78°C and the resulting solution was stirred at this temperature for 1 hour. A solution of (2R,3S,4S)-2,3,4-tris-benzyloxy-5-hydroxy-6-(4-methoxy-benzyloxy)-5-(4-methoxy-benzyloxymethyl)-hexanoic acid methoxy-methyl-amide I-1g (725.0 mg, 0.930 mmol) in anhydrous tetrahydrofuran (1.5 mL) was then added dropwise very slowly (0.9 ml/hour) and the resulting mixture was stirred at -78°C for an additional hour before warming to 10°C over 12 hours (put in a deep Dewar covered with aluminum foil to maintain cold temperature). The reaction was diluted with diethyl ether and quenched by the dropwise addition of aqueous 1 N hydrochloric acid solution. The resulting biphasic mixture was stirred at room temperature for 15 minutes. The organic phase was separated, washed with brine, dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The crude residue was purified by flash chromatography over silica gel (eluting with a gradient of 10 to 40% ethyl acetate in heptane) to afford the product as a mixture of isomers (535 mg, 59 % yield).
  • ((2S,3S)-2,3,4-tris-benzyloxy-5-{4-fluoro-3-[4-(oxetan-3-yloxy)-benzyl]-phenyl}-6,8-dioxa-bicyclo[3.2.1]oct-1-yl]-methanol (I-13k)
  • Figure imgb0033
  • To a solution of (4S,5S)-3,4,5-Tris-benzyloxy-2-{4-fluoro-3-[4-(oxetan-3-yloxy)-benzyl]-phenyl}-6,6-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-pyran-2-ol I-13i (535 mg, 0.548 mmol) in dichloromethane (2.0 mL) was added anisole (0.480 mL, 4.38 mmol) followed by a 20% solution of trifluoroacetic acid in dichloromethane (8.0 mL) at room temperature under nitrogen. After stirring for 1 hour, the reaction mixture was concentrated under reduced pressure. The crude residue was purified by flash chromatography over silica gel (eluting with a gradient of 10 to 30% ethyl acetate in heptane) to afford the product as a mixture of isomers (300 mg, 76% yield).
  • (4S,5S)-3,4,5-tris-benzyloxy-2-{4-chloro-3-{4-(oxetan-3-yloxy)-benzyl]-phenyl}-6,6-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-pyran-2-ol (I-14i):
  • Figure imgb0034
  • n-Butyl lithium (0.97 mL, 2.5 M/hexanes, 3.15 equivalents) was added dropwise (1 drop every 5 seconds) to an oxygen degassed solution (placed in a pre dried Biotage microwave vial 10-20 mL sealed with its cap and placed under a positive stream of nitrogen gas) of 3-(4-(5-bromo-2-chlorobenzyl)phenoxy)oxetane (824 mg, 2.95 equivalents) in anhydrous tetrahydrofuran (2.7 mL) at -78°C and the resulting solution was stirred at this temperature for an additional hour. A solution of (2R,3S,4S)-2,3,4-tris-benzyloxy-5-hydroxy-6-(4-methoxy-benzyloxy)-5-(4-methoxy-benzyloxymethyl)-hexanoic acid methoxy-methyl-amide (I-1g) (616 mg) in anhydrous tetrahydrofuran (1.35 mL) was then added dropwise over 1.5 hours using a syringe pump and the resulting mixture was stirred at - 78°C for 1 hour before being allowed to warm to -20°C over 14 hours (put in a deep Dewar covered with aluminum foil to maintain cold temperature; size of Dewar: external diameter 10 cm, internal diameter 8 cm, height 9 cm). Diethyl ether was added and the reaction was quenched by dropwise addition of aqueous 1 M hydrochloric acid solution. The resulting biphasic mixture was stirred at room temperature for 15 minutes. The organic phase was separated, dried over sodium sulfate, filtered and concentrated. Chromatography over silica gel using a gradient of 0-50% ethyl acetate in heptane gave the product as a mixture of isomers (563 mg, 72% yield).
  • ((2S, 3S)-2,3,4-Tris-benzyloxy-5-{4-chloro-3-[4-(oxetan-3-yloxy)-benzyl]-phenyl}-6,8-dioxa-bicyclo[3.2.1]oct-1-yl)-methanol (I-14k):
  • Figure imgb0035
  • To a solution of the intermediate (4S,5S)-3,4,5-tris-benzyloxy-2-{4-chloro-3-[4-(oxetan-3-yloxy)-benzyl]-phenyl}-6,6-bis-(4-methoxy-benzyloxymethyl)-tetrahydro-pyran-2-ol I-14i (282 mg) in dichloromethane (2.84 mL) was added anisole (200 microL, ~7 equivalents) followed by 3.07 mL of a solution of 20% trifluoroacetic acid in dichloromethane and the resulting mixture was stirred at room temperature for 1.5 hours. The mixture was concentrated and the crude was chromatographed over silica gel using a gradient of 10 to 50% ethyl acetate in heptane to afford the product as a mixture of isomers (186 mg, 89% yield).
  • Example 1 (1S,2S, 3S,4R, 5S)-1-hydroxymethyl-5-[3-(4-methoxy-benzyl)-4-methyl-phenyl]-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (1A) and (1S,2S,3S,4S,5S)-1-hydroxymethyl-5-[3-(4-methoxy-benzyl)-4-methyl-phenyl]-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (1B):
  • Figure imgb0036
  • To a solution of {(2S,3S)-2,3,4-tris-benzyloxy-5-[3-(4-methoxy-benzyl)-4-methyl-phenyl]-6,8-dioxa-bicyclo[3.2.1]oct-1-yl}-methanol (I-1k: 236 mg) in ethanol/tetrahydrofuran (7 mL, 4/1 volume) was added successively formic acid (270 microL, 19 equivalents) and palladium black (150 mg, 4 equivalents) and the resulting mixture was stirred at room temperature for 3 hours. The palladium was filtered and the crude mixture obtained after evaporation of solvent was purified by chromatography over silica gel eluting with a gradient of 85 to 100% ethyl acetate in heptane. The mixture of products obtained was purified by HPLC preparative.
  • HPLC preparative method: reverse phase C18 phenomenex column Luna 5 micrometer 150 x 21.20 mm, 20 mL/minute, gradient of acetonitrile/0.1 % formic acid:water/0.1 % formic acid; 20 to 60% of acetonitrile/0.1 % formic acid over 20 minutes. UV detection: 254 nm. The HPLC indicated a ratio of diastereoisomers of 3:1 (1A:1B).
  • 1A: (55 mg, 39% yield); Rt = 10.9 minutes; the fractions containing the product were concentrated under reduced pressure. The crude material was precipitated from ethyl acetate and heptane. The resulting white solid was washed with heptane 2 times and dried under reduced pressure. MS (LCMS) 403.3 (M+H+; positive mode) 447.3 (M+HCO2 -, negative mode).
  • 1H NMR (400 MHz, methanol-d4) delta 7.33 (d, 1H, J = 1.6 Hz), 7.30 (dd, 1H, J = 7.6 and 1.6 Hz), 7.10 (d, 1H, J = 7.6 Hz), 7.02-6.98 (m, 2H), 6.79-6.75 (m, 2H), 4.13 (d, 1H, J = 7.4 Hz), 3.90 (s, 2H), 3.82 (d, 1H, J = 12.5 Hz), 3.77 (dd, 1H, J = 8.2 and 1.2 Hz), 3.72 (s, 3H), 3.66 (d, 1H, J = 12.5 Hz), 3.65 (t, 1H, J = 8.0 Hz), 3.59 (d, 1H, J = 7.8 Hz), 3.58 (dd, 1H, J = 7.5 and 1.5 Hz), 2.16 (s, 3H). HRMS calculated for C22H27O7 (M+H+) 403.1751, found 403.1737.
  • 1B: (20 mg, 14% yield); Rt = 11.5 minutes; the fractions containing the product were concentrated under reduced pressure. The crude material was precipitated from ethyl acetate and heptane. The resulting white solid was washed with heptane 2 times and dried under reduced pressure. MS (LCMS) 403 (M+H+; positive mode) 447 (M+HCO2 -, negative mode).
  • 1H NMR (400 MHz, methanol-d 4) delta 7.38 (d, 1H, J = 1.8 Hz) 7.33 (dd, 1H, J = 7.9 and 1.8 Hz), 7.10 (d, 1H, J = 7.9 Hz), 7.02-6.97 (m, 2H), 6.79-6.74 (m, 2H), 4.02 (d, 1H, J = 7.4 Hz), 3.93 (t, 1H, J=2.2 Hz), 3.91 (br. s, 2H), 3.88 (d, 1H, J = 12.5 Hz), 3.84 (d, 2H, J = 2.4 Hz), 3.75 (d, 1H, J = 12.5 Hz), 3.71 (s, 3H), 3.49 (d, 1H, J = 7.4 Hz), 2.16 (s, 3H).
  • Example 2 (1S,2S, 3S,4R, 5S)-5-[3-(4-ethoxy-benzyl)-4-methyl-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (2A) and (1S,2S,3S,4S,5S)-5-[3-(4-ethoxy-benzyl)-4-methyl-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2.3.4-triol (2B)
  • Figure imgb0037
  • To a solution of {(25,3S)-2,3,4-tris-benzyloxy-5-[3-(4-ethoxy-benzyl)-4-methyl-phenyl]-6,8-dioxa-bicyclo[3.2.1]oct-1-yl}-methanol (I-2k: 85 mg, 0.12 mmol) in ethanol/tetrahydrofuran (7 mL, ~4/1 volume) was added successively formic acid (95 microL, 19 equivalents) and palladium black (53 mg, 4 equivalents) and the resulting mixture was stirred at room temperature for 3 hours. The palladium was filtered and the crude mixture obtained after evaporation of solvent was purified by HPLC preparative.
  • HPLC preparative method: reverse phase C18 phenomenex column Luna 5 micrometer 150 x 21.20 mm, 20 mL/minute, gradient of acetonitrile/0.1 % formic acid:water/0.1 % formic acid; 20 to 60% of acetonitrile/0.1 % formic acid over 20 minutes. UV detection: 254 nm. The HPLC indicated a ratio of diastereoisomers of 4:1 (2A:2B).
  • 2A: (20 mg; 38% yield) Rt = 12.7 minutes; the fractions containing the product were concentrated under reduced pressure. The crude material was precipitated from ethyl acetate and heptane. The resulting white solid was washed with heptane 2 times and dried under reduced pressure.
  • MS (LCMS) 417.3 (M+H+; positive mode); 461.4 (M+HCO2 -; negative mode). 1H NMR (400 MHz, methanol-d4) delta ppm 1.34 (t, J=6.9 Hz, 3 H), 2.18 (s, 3 H), 3.60 (d, J=8 Hz, 2 H), 3.66 (t, J=8 Hz, 1 H), 3.68 (d, J=12.5 Hz, 1 H), 3.78 (d, 1 H, J= 8.8 Hz), 3.84 (d, J=12.4 Hz, 1 H), 3.92 (s, 2 H), 3.97 (q, J=7 Hz, 2 H), 4.15 (d, J=7.5 Hz, 1 H), 6.77 (m, 2 H), 7.00 (m, 2 H), 7.12 (d, J=7.7 Hz, 1 H), 7.31 (dd, J=7.9 and 1.4 Hz, 1 H), 7.34 (s, 1 H).
  • 2B: (5 mg; 9% yield) Rt = 13.2; minutes the fractions containing the product were concentrated under reduced pressure. The crude material was precipitated from ethyl acetate and heptane. The resulting white solid was washed with heptane 2 times and dried under reduced pressure.
  • MS (LCMS) 417.3 (M+H+; positive mode); 461.4 (M+HCO2 -; negative mode). 1H NMR (400 MHz, methanol-d 4) delta ppm 1.34 (t, J=6.9 Hz, 3 H), 2.18 (s, 3 H), 3.52 (d, 1H, J= 7.4 Hz), 3.77 (d, J=12.5 Hz, 1 H), 4.00-3.84 (m, 8 H), 4.04 (d, J=7.4 Hz, 1 H), 6.79-6.75 (m, 2 H), 7.03-6.98 (m, 2 H), 7.12 (d, J=7.9 Hz, 1 H), 7.35 (dd, J=7.7 and 1.9 Hz, 1 H), 7.39 (d, J = 1.9 Hz, 1H).
  • Example 3 (1S,2S,3S, 4R,5S)-5-[4-chloro-3-(4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3-2-1]octane-2,3,4-triol (3A) and (1S,2S,3S,4S,5S)-5-[4-chloro-3-[4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2.3.4-triol (3B):
  • Figure imgb0038
  • To a solution of {(2S,3S)-2,3,4-tris-benzyloxy-5-[4-chloro-3-(4-methoxy-benzyl)-phenyl]-6,8-dioxa-bicyclo[3.2.1]oct-1-yl}-methanol (I-3k: 229 mg) in ethanol/tetrahydrofuran (7 mL, 4/1 volume) was added successively formic acid (270 microL, 20 equivalents) and palladium black (140 mg, 4 equivalents) and the resulting mixture was stirred at room temperature. After 1 hour, additional formic acid (270 microL, 20 equivalents) and palladium black (140 mg, 4 equivalents) were added and the mixture was allowed to stir for an additional hour at room temperature. The palladium was filtered and the crude mixture obtained after evaporation of solvent was purified by HPLC preparative.
  • HPLC preparative method: reverse phase C18 phenomenex column Luna 5 micrometer 150 x 21.20 mm, 20 mL/minutes, gradient of acetonitrile/0.1 % formic acid:water/0,1 % formic acid; 20 to 60% of acetonitrile/0.1 %formic acid over 20 minutes. UV detection: 254 nm. The HPLC indicated a ratio of diastereoisomers of 1.4:1 (3A:3B).
  • 3A: (50 mg; 36% yield) Rt = 12.1 minutes; the fractions containing the product were concentrated under reduced pressure. The crude material was precipitated from ethyl acetate and heptane. The resulting white solid was washed with heptane 2 times and concentrated under reduced pressure.
  • MS (LCMS) 423.3 (M+H+; positive mode); 467.3 (M+HCO2 -; negative mode). 1H NMR (400 MHz, methanol-d4) delta 7.43 (s, 1H), 7.38-7.30 (m, 2H), 7.08 (d, 2H), 6.79 (d, 2H), 4.12 (d, 1H, J = 7.5 Hz), 4.01 (s, 2H), 3.81 (d, 1H, J=12.5 Hz), 3.75 (d, 1H, J = 8.4 Hz), 3.73 (s, 3H), 3.66 (d, 1H, J = 11.7 Hz), 3.63 (t, 1H, J=8.2 Hz), 3.57 (d, 1H, J=7.4 Hz), 3.52 (d, 1H, J = 7.8 Hz). HRMS calculated for C21H24O7Cl (M+H+) 423.1205, found 423.1192.
  • 3B: (37 mg; 27% yield) Rt = 12.8 minutes; the fractions containing the product were concentrated under reduced pressure. The crude material was precipitated from ethyl acetate and heptane. The resulting white solid was washed with heptane 2 times and concentrated under reduced pressure.
  • MS (LCMS) 423.3 (M+H+; positive mode) 467.3 (M+HCO2 -, negative mode). 1H NMR (400 MHz, methanol-d4) delta 7.50 (d, 1H, J = 1.9 Hz) 7.42 (dd, 1H, J = 8.3 and 1.9 Hz), 7.35 (d, 1H, J = 8.3 Hz), 7.12-7.07 (m, 2H), 6.83-6.78 (m, 2H), 4.06-4.01 (m, 3H), 3.91-3.83 (m, 4H), 3.78-3.72 (m, 4H), 3.51 (d, 1H, J = 7.5 Hz).
  • Example 4 (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (4A) and (1S,2S,3S,4S,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (4B):
  • Figure imgb0039
  • To a solution of {(2S,3S)-2,3,4-tris-benzyloxy-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-6,8-dioxa-bicyclo[3.2.1]oct-1-yl}-methanol (I-4k: 335 mg) in ethanol/tetrahydrofuran (10 mL, 4/1 volume) was added successively formic acid (420 microL, 22 equivalents) and palladium black (208 mg, 4 equivalents) and the resulting mixture was stirred at room temperature. After 1 hour, additional formic acid (420 microL, 22 equivalents) and palladium black (208 mg, 4 equivalents) were added and the mixture was allowed to stir for an additional hour at room temperature. The palladium was filtered and the crude mixture obtained after evaporation of solvent was purified by HPLC preparative.
  • HPLC preparative: reverse phase C18 Gemini column 5 micrometer 30 x 100 mm, 40 mL/minute, gradient of acetonitrile/0.1 % formic acid : water/0.1 % formic acid; 25 to 50% of acetonitrile/0.1 % formic acid over 18 minutes; UV detection: 220 nm. The HPLC indicated a ratio of diastereomers of 1.1:1 (4A:4B).
  • 4A: (60 mg, 29% yield); Rt = 12.4 minutes; the fractions containing the product were concentrated under reduced pressure. The crude material was precipitated from ethyl acetate and heptane. The resulting white solid was washed with heptane 2 times and dried under reduced pressure.
  • MS (LCMS) 437.3 (M+H+; positive mode); 481.3 (M+HCO2 -; negative mode). 1H NMR (400 MHz, methanol-d4) delta 7.43 (d, 1H, J = 1.9 Hz), 7.36 (dd, 1H, J = 8.3 and 2 Hz), 7.32 (d, 1H, J = 8.3 Hz), 7.08-7.04 (m, 2H), 6.79-6.75 (m, 2H), 4.12 (d, 1H, J = 7.5 Hz), 4.00 (s, 2H), 3.96 (q, 2H, J = 7.0 Hz), 3.81 (d, 1H, J = 12.5 Hz), 3.75 (dd, 1H, J = 8.3 and 1.3 Hz), 3.65 (d, 1H, J = 12.5 Hz), 3.63 (t, 1H, J = 8.2 Hz), 3.57 (dd, 1H, J = 7.5 and 1.3 Hz), 3.52 (d, 1H, J = 8.0 Hz), 1.33 (t, 3H, J = 6.9 Hz). HRMS calculated for C22H26O7Cl (M+H+) 437.1361, found 437.1360.
  • 4B: (30 mg, 15% yield); Rt = 13.2 minutes; the fractions containing the product were concentrated under reduced pressure. The crude material was precipitated from ethyl acetate and heptane. The resulting white solid was washed with heptane 2 times and dried under reduced pressure.
  • MS (LCMS) 437.3 (M+H+; positive mode) 481.3 (M+HCO2 -, negative mode). 1H NMR (400 MHz, methanol-d 4) delta 7.48 (d, 1H, J = 1.9 Hz) 7.40 (dd, 1H, J = 8.1 and 1.9 Hz), 7.32 (d, 1H, J = 8.3 Hz), 7.08-7.03 (m, 2H), 6.80-6.74 (m, 2H), 4.04-3.99 (m, 3H), 3.95 (q, 2H, J = 7 Hz), 3.89-3.81 (m, 4H), 3.73 (d, 1H, J = 12.5 Hz), 3.49 (d, 1H, J = 7.3 Hz), 1.32 (t, 3H, J = 7 Hz). HRMS calculated for C22H26O7Cl (M+H+) 437.1361, found 437.1358.
  • Example 5 (1S,2S,3S,4R,5S)-5-[4-fluoro-3-(4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2.3.4-triol (5A) and (1S,2S,3S,4S,5S)-5-[4-fluoro-3-(4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (5B):
  • Figure imgb0040
  • To a solution of {(2S,3S)-2,3,4-Tris-benzyloxy-5-[4-fluoro-3-(4-methoxy-benzyl)-phenyl]-6,8-dioxa-bicyclo[3.2.1]oct-1-yl}-methanol (115 mg, 0.170 mmol) in a 4:1 solution of ethanol/tetrahydrofuran (10 mL) was added successively formic acid (137 microL, 3.42 mmol) and palladium black (73 mg, 0.687 mmol). The resulting mixture was stirred at room temperature. After 3 hours, additional formic acid (137 microL, 3.42 mmol) and palladium black (73 mg, 0.687 mmol) were added. After 18 hours, the reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The resulting crude residue was purified by flash chromatography over silica gel (eluting with a gradient of 0 to 15% methanol in dichloromethane) to afford 64 mg of a white solid. The mixture of isomers was purified by preparative HPLC.
  • HPLC preparative method: reverse phase C18 phenomenex column Luna 5 micrometer 150 x 21.20 mm, 20 mL/minute, gradient of acetonitrile/0.1 % formic acid:water/0.1 % formic acid; 20 to 80% of acetonitrile/0.1 % formic acid over 20 minutes). UV detection: 254 nm. The HPLC indicated a ratio of diastereomers of 1:1 (5A:5B).
  • 5A: (6 mg; 9% yield) Rt = 8.5 minutes; the fractions containing the product were concentrated under reduced pressure. The crude material was precipitated from ethyl acetate and heptane. The resulting white solid was washed with heptane 2 times and dried under reduced pressure.
  • 1H NMR (400 MHz, methanol-d4) delta ppm 3.55 (d, J=7.8 Hz, 1 H), 3.58 (dd, J=7.5, 1.2 Hz, 1 H), 3.64 (t, J=8.2 Hz, 1 H), 3.67 (d, J=12.5 Hz, 1 H), 3.74 (s, 3 H), 3.77 (dd, J=8.3, 1.2 Hz, 1 H), 3.83 (d, J=12.5 Hz, 1 H), 3.91 (s, 2 H), 4.14 (d, J=7.4 Hz, 1 H), 6.76 - 6.84 (m, 2 H), 7.02 (dd, J=9.9, 8.3 Hz, 1 H), 7.09 - 7.13 (m, 2 H), 7.37 - 7.44 (m, 2 H); MS: 407.4 (M+ H+; positive mode); 451.3 (M+HCO2 -; negative mode)
  • 5B: (12 mg; 17% yield) Rt = 9 minutes; the fractions containing the product were concentrated under reduced pressure. The crude material was precipitated from ethyl acetate and heptane. The resulting white solid was washed with heptane 2 times and dried under reduced pressure.
  • 1H NMR (400 MHz, methanol-d 4) delta ppm 3.51 (d, J=7.4 Hz, 1 H), 3.74 (s, 3 H), 3.75 (d, 1H, J = 13 Hz), 3.83 - 3.93 (m, 6 H), 4.03 (d, J=7.4 Hz, 1H), 6.78 - 6.82 (m, 2 H), 7.02 (dd, J=9.9, 8.5 Hz, 1 H), 7.09 - 7.13 (m, 2 H), 7.42 - 7.49 (m, 2 H); MS: 407.4 (M+ H+; positive mode); 451.3 (M+HCO2 -; negative mode)
  • Example 6 2-(4-methoxybenzyl)-4-((1S,2S,3S,4R,5S)-2,3,4-trihydroxy-1-(hydroxymethyl)-6,8-dioxa-bicyclo[3,2,1]oct-5-yl)-benzonitrile (6A):
  • Figure imgb0041
  • n-Butyl lithium (1.04 mL, 2.6 mmol, 2.5 M in hexane) was added to a solution of isopropyl magnesium bromide (1.27 mL, 1.27 mmol, 1 M in tetrahydrofuran) at 0 °C. After being stirred for 30 minutes, the resulting mixture was cooled to -78°C and a solution of 4-bromo-2-(4-ethoxy-benzyl)-benzonitrile (380 mg, 1.20 mmol) in anhydrous tetrahydrofuran (1 mL) was added. The greenish mixture was stirred for 1 hour at -78°C and a solution of (2R,3S,4S)-2,3,4,6-tetrakis-benzyloxy-5-benzyloxymethyl-5-hydroxy-hexanoic acid methoxy-methyl-amide (I-6g) (700 mg, 0.972 mmol) in anhydrous tetrahydrofuran (2 mL) was added very slowly (over 20 minutes, 1 drop every 5 seconds). The solution was stirred at -78°C for 1 hour and slowly warmed up to room temperature over 3 hours. The reaction was quenched by dropwise addition of aqueous 1 M hydrochloric acid solution and then diluted with ethyl acetate. The resulting biphasic mixture was stirred at room temperature for 15 minutes. The organic phase was separated, washed with brine, dried over magnesium sulfate, filtered and concentrated, affording the crude product. The crude product was purified by flash chromatography over silica gel eluting with a gradient of 0 to 20% ethyl acetate in heptane affording the desired intermediate 2-(4-Ethoxy-benzyl)-4-((4S,5S)-3,4,5-tris-benzyloxy-6,6-bis-benzyloxymethyl-2-hydroxy-tetrahydro-pyran-2-yl)-benzonitrile (300 mg; 34% yield). MS 918.8 (M+Na+, positive mode).
  • Boron trichloride (4.18 mL, 4.18 mmol, 1 M solution in hexane) was added to the solution of the above intermediate (250 mg, 0.279 mmol) in CH2Cl2 (2 mL) at -78 °C. The mixture was stirred at -78 °C for 10 minutes and then warmed up to room temperature overnight. The mixture was quenched with water (10 mL) and extracted with ethyl acetate (50 mL). The organic layer was dried over sodium sulfate and evaporated to dryness. Purification by flash chromatography over silica gel (eluting with methanol in dichloromethane : 1 to 9 in volume) gave the desired intermediate 2-(4-Hydroxy-benzyl)-4-((1S,2S,3S,4R,5S)-2,3,4-trihydroxy-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]oct-5-yl)-benzonitrile (35 mg, 30% yield).
  • Potassium carbonate (28 mg, 0.2 mmol) was added to a solution of the above intermediate (34 mg, 0.077 mmol) in acetone (0.4 mL), followed by iodomethane (7 microL, 0.11 mmol) at room temperature. The mixture was stirred at 45°C overnight. The mixture was diluted with ethyl acetate (60 mL) and washed with water. The organic layer was dried over sodium sulfate and evaporated to dryness. Purification by preparativeThin Layer Chromatography over silica gel (eluting with methanol in dichloromethane : 1 to 9 in volume) allowed isolation of desired product 6A (18 mg; 57% yield).
  • 1H NMR (400 MHz, methanol-d 4) delta 7.69 (d, J = 8 Hz, 1H), 7.61 (s, 1H), 7.56 (d, J = 8 Hz, 1H), 7.19-7.14 (m, 2H), 6.87-6.82 (m, 2H), 4.18 (d, J = 7.6 Hz, 1H), 4.14 (s, 2H), 3.86 (d, J = 12.7 Hz, 1H); 3.81 (d, J = 8.3 Hz, 1H), 3.76 (s, 3H), 3.69 (d, J = 12.5 Hz, 1H), 3.67 (t, J=8.1 Hz, 1H), 3.61 (d, J = 7.6 Hz, 1H), 3.54 (d, J = 8 Hz, 1H); MS 458.4 (M+HCO2 -; negative mode).
  • Example 7 2-(4-ethoxybenzyl)-4-((1S,2S,3S,4R,5S)-2,3,4-trihydroxy-1-(hydroxymethyl)-6,8-dioxa-bicyclo[3.2.1]oct-5-yl)-benzonitrile(7A):
  • Figure imgb0042
  • Potassium carbonate (8 mg, 0.058 mmol) was added to a solution of intermediate 2-(4-hydroxy-benzyl)-4-((1S,2S,3S,4R,5S)-2,3,4-trihydroxy-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]oct-5-yl)-benzonitrile (see example 6; 8.9 mg, 0.022 mmol) in acetone (0.4 mL), followed by iodoethane (4 microL, 0.044 mmol) at room temperature. The mixture was stirred at 45°C overnight. The mixture was diluted with ethyl acetate (60 mL) and washed with water. The organic layer was dried over sodium sulfate and evaporated to dryness. Purification by preparative Thin Layer Chromatography over silica gel (eluting with methanol in dichloromethane : 1 to 9 in volume) allowed isolation of desired product 7A (2.4 mg; 26% yield).
  • 1H NMR (methanol-d4) delta 7.69 (d, J = 8.0 Hz, 1H), 7.61 (d, J = 1.5 Hz, 1H), 7.56 (dd, J = 8.0, 1.5 Hz, 1H), 7.17-7.13 (m, 2H), 6.86-6.81 (m, 2H), 4.18 (d, J = 7.5 Hz, 1H), 4.14 (s, 2H), 4.01 (q, J = 7.0 Hz, 2H); 3.86 (d, J = 12.5 Hz, 1H); 3.80 (dd, J = 8.0 and 1.2 Hz, 1H), 3.70 (d, J =11.7 Hz, 1H), 3.67 (t, J = 8.0 Hz, 1H), 3.61 (dd, J = 7.5 and 1.2 Hz, 1H), 3.54 (d, J = 7.8 Hz, 1H), 1.37 (t, J = 7.0 Hz, 3H); MS 472.1 (M+HCO2 -; negative mode).
  • Example 8 illustrates the preparation of a crystalline derivative of the compound of Example 3B in order to confirm the structure and stereochemistry of Example 3B.
  • Example 8 Per 4-bromobenzoylation of (1S,2S,3S,4S,5S)-5-[4-chloro-3-(4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (3B) to give (8A):
  • Figure imgb0043
  • To a solution of (1S,2S,3S,4S,5S)-5-[4-chloro-3-(4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (3B) (11 mg, 0.026 mmol) in anhydrous tetrahydrofuran (600 microL) were added at room temperature N,N-diisopropylethylamine (32 microL, 7 equivalents) and 4-dimethylaminopyridine (3 mg, 0.9 equivalents) followed by para-bromobenzoyl chloride (35 mg, 6 equivalents) and the resulting mixture was stirred at room temperature for 62 hours. Ethyl acetate and water were added and the organic phase was successively washed with 0.5M aqueous hydrochloric acid solution and brine. The organic phase was dried over magnesium sulfate, filtered, concentrated and the crude was purified by flash chromatography over silica gel eluting with a gradient of 15 to 30% ethyl acetate in heptane to afford 27 mg of product (90% yield).
  • 1H NMR (400 MHz, chloroform-d) delta 7.82 (m, 2H), 7.74-7.64 (m, 4H), 7.58-7.46 (m, 8H), 7.42-7.34 (m, 4H), 7.29 (d, 1H, J = 8.3 Hz), 6.89 (m, 2H), 6.63 (m, 2H), 6.04 (dd, 1H, J = 9.6 and 1 Hz), 5.98 (dd, 1H, J = 9.6 and 4.4 Hz), 5.89 (d, 1H, J = 4.4 Hz), 4.70 (d, 1H, J = 12.4 Hz), 4.65 (d, 1H, J = 12.4 Hz), 4.60 (d, 1H, J = 8 Hz), 3.98-3.88 (m, 3H), 3.73 (s, 3H).
  • Single crystals were obtained by vapor diffusion techniques using heptane and ethyl acetate as solvents. Melting point = 191° C. Single Crystal X-Ray Analysis. A representative crystal was surveyed and a 1 Å data set (maximum sin Θ/λ=0.5) was collected on a Bruker APEX II/R diffractometer. Friedel pairs were collected in order to facilitate the determination of the absolute configuration. Atomic scattering factors were taken from the International Tables for Crystallography. See, International Tables for Crystallography, Vol. C, pp. 219, 500, Kluwer Academic Publishers,1992. All crystallographic calculations were facilitated by the SHELXTL system. See, SHELXTL, Version 5.1, Bruker AXS, (1997). All diffractometer data were collected at room temperature. Pertinent crystal, data collection, and refinement are summarized in Table 1 below. Table 1
    Crystal data and structure refinement for Example 8A.
    Empirical formula C49H35O11Br4Cl
    Formula weight 1154.86
    Temperature 296(2) K
    Wavelength 1.54178 Å
    rystal system Monoclinic
    Space group C2
    Unit cell dimensions a = 23.7485(6) A α=90°.
    b = 6.3175(2) A β=104.4910(10)°.
    c = 32.3167(8) Å γ=90°.
    Volume 4694.3(2) Å3
    Z 4
    Density (calculated) 1.634 Mg/m3
    Absorption coefficient 5.216 mm-1
    F(000) 2296
    Crystal size 0.12 x 0.03 x 0.02 mm3
    Theta range for data collection 3.75 to 50.43°.
    Reflections collected 8339
    Independent reflections 3932 [R(int) = 0.0491]
    Completeness to theta = 50.43° 89.7 %
    Absorption correction Empirical Absorption Correction
    Refinement method Full-matrix least-squares on F2
    Data / restraints / parameters 3932/1/587
    Goodness-of-fit on F2 0.967
    Final R indices [I>2sigma(I)] R1 = 0.0371, wR2 = 0.0854
    Absolute structure parameter -0.03(2)
    Extinction coefficient 0.00011 (3)
    Largest diff. peak and hole 0.297 and -0.294 e.Å-3
  • A trial structure was obtained by direct methods. This trial structure refined routinely. Hydrogen positions were calculated wherever possible. The methyl hydrogens were located by difference Fourier techniques and then idealized. The hydrogen parameters were added to the structure factor calculations but were not refined. The shifts calculated in the final cycles of least squares refinement were all less than 0.1 of the corresponding standard deviations. The final R-index was 3.71 %. A final difference Fourier revealed no missing or misplaced electron density. The refined structure was plotted using the SHELXTL plotting package (Figure 1). The absolute configuration was determined by the method of Flack. See, Flack, H.D., Acta Crystallogr., A39, 876, (1983).
  • Example 9 illustrates the preparation of a crystalline derivative of the compound of Example 4A in order to confirm the structure and stereochemistry of Example 4A.
  • Example 9 Per 4-nitrobenzoylation of (1S,2S,3S,4R,5S)-5[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (4A) to give (9A):
  • Figure imgb0044
  • To a solution of (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (4A: 10.6 mg, 0.024 mmol) in anhydrous tetrahydrofuran (300 microL) cooled at 0°C were added N,N-diisopropylethylamine (30 microL, 7 equivalents) and 4-dimethylaminopyridine (3 mg, 1 equivalents) followed by para-nitrobenzoyl chloride (27 mg, 6 equivalents) and the resulting mixture was stirred at 60° C for 6 hours. The mixture was cooled to room temperature, ethyl acetate and water were added and the organic phase was successively washed with 0.5M aqueous hydrochloric acid solution and brine. The organic phase was dried over magnesium sulfate, filtered, concentrated and the crude was purified by flash chromatography over silica gel eluting with a gradient of 10 to 50% ethyl acetate in heptane to afford 18 mg of product (73% yield).
  • 1H NMR (400 MHz, chloroform-d) delta 8.33 (m, 2H), 8.28-8.12 (m, 8H), 8.07 (m, 2H), 8.00 (m, 2H), 7.91 (m, 2H), 7.45-7.40 (m, 2H), 7.34 (d, 1H, J = 8.2 Hz), 6.87 (m, 2H), 6.64 (m, 2H), 6.13 (d, 1H, J = 8.6 Hz), 6.06 (t, 1H, J = 8.3 Hz), 5.86 (d, 1H, J = 8.1 Hz), 4.81 (d, 1H, J = 8.3 Hz), 4.75 (d, 1H, J = 12.7 Hz), 4.60 (d, 1H, J = 12.8 Hz), 4.06 (d, 1H, J = 8.5 Hz), 3.98-3.90 (m, 4H), 1.39 (t, 3H, J = 7 Hz).
  • Single crystals were obtained by slow recrystallization from acetonitrile/ isopropanol as solvents. Melting point = 211°C. A representative crystal was surveyed and a 0.88 Å data set (maximum sin Θ/λ=0.57) was collected on a Bruker APEX II/R diffractometer. Friedel pairs were collected in order to facilitate the determination of the absolute configuration. Atomic scattering factors were taken from the International Tables for Crystallography. See, International Tables for Crystallography, Vol. C, pp. 219, 500, Kluwer Academic Publishers,1992. All crystallographic calculations were facilitated by the SHELXTL system. See, SHELXTL, Version 5.1, Bruker AXS, (1997). All diffractometer data were collected at room temperature. Pertinent crystal, data collection, and refinement are summarized in Table 2 below. Table 2
    Crystal data and structure refinement for Example 9A.
    Empirical formula C50H37N4O19Cl
    Formula weight 1033.29
    Temperature 296(2) K
    Wavelength 1.54178 Å
    Crystal system Monoclinic
    Space group P2(1)
    Unit cell dimensions a = 17.5050(4) Å α=90°.
    b = 6.2303(2) Å β=97.6580(10)°.
    c = 21.9545(5) Å γ=90°.
    Volume 2373.03(11) Å3
    Z 2
    Density (calculated) 1.446 Mg/m3
    Absorption coefficient 1.452 mm-1
    F(000) 1068
    Crystal size 0.18 x 0.02 x 0.01 mm3
    Theta range for data collection 2.55 to 61.76°.
    Reflections collected 8972
    Independent reflections 5062 [R(int) = 0.0236]
    Completeness to theta = 61.76° 85.8 %
    Absorption correction Empirical Absorption Correction
    Max. and min. transmission 0.9856 and 0.7801
    Refinement method Full-matrix least-squares on F2
    Data / restraints / parameters 5062/1/668
    Goodness-of-fit on F2 1.009
    Final R indices [I>2sigma(I)] R1 = 0.0436, wR2 = 0.1090
    Absolute structure parameter 0.02(3)
    Extinction coefficient 0.0015(2)
    Largest diff. peak and hole 0.217 and -0.173 e.Å-3
  • A trial structure was obtained by direct methods. This trial structure refined routinely. Hydrogen positions were calculated wherever possible. The methyl hydrogens were located by difference Fourier techniques and then idealized. The hydrogen parameters were added to the structure factor calculations but were not refined. The shifts calculated in the final cycles of least squares refinement were all less than 0.1 of the corresponding standard deviations. The final R-index was 4.36%. A final difference Fourier revealed no missing or misplaced electron density.
  • The refined structure was plotted using the SHELXTL plotting package (Figure 2). The absolute configuration was determined by the method of Flack See, Flack, H.D., Acta Crystallogr., A39, 876, (1983).
  • Example 10 (1S,2S,3S,4R,5S)-5-[3-(4-ethoxy-benzyl)-4-fluoro-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]loctane-2,3,4-triol(10A) and (1S,2S,3S,4S,5S)-5-[3-(4-ethoxy-benzyl)-4-fluoro-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (10B)
  • Figure imgb0045
  • To a solution of {(2S,3S)-2,3,4-tris-benzyloxy-5-[3-(4-ethoxy-benzyl)-4-fluoro-phenyl]-6,8-dioxa-bicyclo[3.2.1]oct-1-yl}-methanol I-10k (80.0 mg, 0.120 mmol) in a 4:1 solution of ethanol/tetrahydrofuran (10 mL) was added successively formic acid (93 microL, 2.32 mmol) and palladium black (62 mg, 0.580 mmol). The resulting mixture was stirred at room temperature. After 3 hours, additional formic acid (93 microL, 2.32 mmol) and palladium black (62 mg, 0.580 mmol) were added. After 5 hours, the reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The resulting crude residue was purified by flash chromatography over silica gel (eluting with a gradient of 0 to 15% methanol in dichloromethane) to afford 35.0 mg of a white solid (mixture of isomers). The mixture of isomers was purified by preparative HPLC.
  • HPLC preparative method: reverse phase C18 Gemini column, 5 micrometer 30x100mm, 40mL/minute flow rate, gradient of acetonitrile/0.1 % formic acid : water/0.1 % formic acid; 25 to 50% acetonitrile/0.1 % formic acid over 18 minutes; UV detection: 220 nm.
  • HPLC analytical method: reverse phase C18 Gemini column, 5 µm 4.6x150mm, 1 mL/minute flow rate, gradient of acetonitrilel0.1 % trifluoroacetic acid : water/0.1 % trifluoroacetic acid; 5 to 100% acetonitrile/0.1 % trifluoroacetic acid over 12 minutes; UV detection: 220 nm.
  • 10A: (2.2 mg, 4.5% yield) Rt = 7 minutes (analytical method); the fractions containing the product were concentrated under reduced pressure.
  • MS (LCMS) 421.4 (M+H+; positive mode) 465.3 (M+HCO2 -, negative mode). 1H NMR (400 MHz, methanol-d4) delta ppm 1.33 (t, J=7.0 Hz, 3 H), 3.53 (d, J=8.0 Hz, 1 H), 3.57 (dd, J=7.5, 1.5 Hz, 1 H), 3.60 - 3.67 (m, 2 H), 3.75 (dd, J=8.3, 1.3 Hz, 1 H), 3.81 (d, J=12.5 Hz, 1 H), 3.89 (s, 2 H), 3.96 (q, J=6.9 Hz, 2 H), 4.12 (d, J=7.4 Hz, 1 H), 6.77 (m, 2 H), 7.00 (dd, J=9.4, 8.2 Hz, 1 H), 7.08 (m, 2 H), 7.36 - 7.41 (m, 2 H).
  • 10B: (1.8 mg, 3.7% yield) Rt = 7.13 minutes (analytical method); the fractions containing the product were concentrated under reduced pressure.
  • MS (LCMS) 421.4 (M+H+; positive mode) 465.3 (M+HCO2 -, negative mode). 1H NMR (400 MHz, methanol-d4) delta ppm 1.34 (t, J=7.0 Hz, 3 H), 3.51 (d, J=7.4 Hz, 1 H), 3.75 (d, 1 H, J = 12.5 Hz), 3.82 - 4.01 (m, 8 H), 4.03 (d, J=7.4 Hz, 1 H), 6.79 (m, 2 H), 7.02 (dd, J=9.8, 8.4 Hz, 1 H), 7.10 (m, 2 H), 7.41 - 7.49 (m, 2 H).
  • Note: after preparative HPLC, the fraction containing these products were concentrated and repurified by flash chromatography over silica gel (eluting with a gradient of 0 to 10% methanol in dichloromethane).
  • Example 11 (1S,2S,3S,4R,5S)-5-{4-fluoro-3-[4-(tetrahydro-furan-3-yloxy)-benzyl]-phenyl}-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (11A)
  • Figure imgb0046
  • To a solution of ((2S,3S)-2,3,4-tris-benzyloxy-5-{4-fluoro-3-[4-(tetrahydro-furan-3-yloxy)-benzyl]-phenyl}-6,8-dioxa-bicyclo[3.2.1]oct-1-yl)-methanol I-11k (160.0 mg, 0.218 mmol) in a 4:1 solution of ethanol/tetrahydrofuran (10 mL) was added successively formic acid (185 microL, 4.64 mmol) and palladium black (148 mg, 1.39 mmol). The resulting mixture was stirred at room temperature. After 3 hours, additional formic acid (185 microL, 4.64 mmol) and palladium black (148 mg, 1.39 mmol) were added. After 5 hours, the reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The resulting crude residue was purified by flash chromatography over silica gel (eluting with a gradient of 0 to 15% methanol in dichloromethane) to afford 100 mg of a white solid (mixture of isomers). The mixture of isomers was purified by preparative HPLC.
  • HPLC preparative method: reverse phase C18 Gemini column, 5 micrometer 30x100mm, 40mL/minute flow rate, gradient of acetonitrile/0.1 % formic acid : water/0.1 % formic acid; 25 to 50% acetonitrile/0.1 % formic acid over 18 minutes; UV detection: 220 nm.
  • HPLC analytical method: reverse phase C18 Gemini column, 5 micrometer 4.6x150mm, 1 mL/minute flow rate, gradient of acetonitrile/0.1 % trifluoroacetic acid : water/0.1 % trifluoroacetic acid; 5 to 100% acetonitrile/0.1 % trifluoroacetic acid over 12 minutes; UV detection: 220 nm.
  • 11A: (19 mg, 19% yield) Rt = 6.43 minutes (analytical method); the fractions containing the product were concentrated under reduced pressure.
  • 1H NMR (400 MHz, methanol-d 4) delta ppm 2.03-2.11 (m, 1 H), 2.15-2.25 (m, 1 H), 3.55 (d, 1 H, J = 8 Hz), 3.59 (dd, 1 H, J = 7.4 and 1 Hz), 3.61 - 3.69 (m, 2 H), 3.77 (dd, J=8.2 and 1 Hz, 1 H), 3.81 - 3.96 (m, 7 H), 4.14 (d, J=7.4 Hz, 1 H), 4.94 - 4.98 (m, 1 H), 6.79 (m, 2 H), 7.02 (dd, J=9.9, 8.5 Hz, 1 H), 7.12 (m, 2 H), 7.37 - 7.45 (m, 2 H).
  • Example 12 (1S,2S,3S,4R,5S)-5-[3-(4-chlorobenzyl)-4-fluorophenyl]-1-hydroxymethyl-6,8-dioxabicyclo(3.2.1]octane-2,3,4-triol (12A) and (1S,2S,3S,4S,5S)-5-[3-(4-chlorobenzyl)-4-fluorophenyl]-1-hydroxymethyl-6,8-dioxabicyclo[3.2.1]octane-2,3,4-triol (12B)
  • Figure imgb0047
  • To a mixture of intermediate I-12k (102 mg) and palladium black (98 mg, 6.1 equivalents) in ethanol/tetrahydrofuran (2 mL, 4/1 volume) was added formic acid (0.9 mL) and the resulting mixture was stirred at room temperature. After 1 hour, additional palladium black (67 mg, 4.2 equivalents) was added and the mixture was allowed to stir for an additional hour at room temperature. The palladium was removed by filtration through Celite® and the filtrate was concentrated, giving the product mixture. This material was combined with a second batch of crude material (prepared from intermediate I-12k (80 mg) following the procedure described above) for purification by preparative HPLC.
  • HPLC preparative conditions: reverse phase C18 Gemini column 5 micrometer 30 x 100 mm, flow rate 40 mL/minute, gradient of acetonitrile/0.1 % formic acid : water/0.1 % formic acid; 25 to 50% of acetonitrile/0.1 % formic acid over 18 minutes), UV detection: 220 nm.
  • HPLC analytical method: reverse phase C18 Gemini column, 5 µm 4.6x150mm, 1 mL/minute flow rate, gradient of acetonitrile/0.1 % trifluoroacetic acid : water/0.1 % trifluoroacetic acid; 5 to 100% acetonitrile/0.1 % trifluoroacetic acid over 12 minutes; UV detection: 220 nm.
  • 12A: (18 mg, 16% yield) Rt = 7.11 minutes (analytical method); MS (LCMS) 411.3 (M+H+; positive mode); 409.2 (M-H+; negative mode). 1H NMR (400 MHz, methanol-d4) delta ppm 7.45-7.42 (m, 2H), 7.25 (d, J = 8.4 Hz, 2H), 7.19 (d, J = 8.4 Hz, 2H), 7.05 (dd, J = 9.6, 9.2 Hz, 1H), 4.15 (d, J=7.6 Hz, 1H), 3.98 (s, 2H), 3.84 (d, J = 12.4 Hz, 1H), 3.78 (dd, J = 8.4, 1.2 Hz, 1H), 3.68 (d, J = 12.8 Hz, 1H), 3.66 (t, J = 8.2 Hz, 1H), 3.60 (dd, J = 7.4, 1.4 Hz, 1H), 3.56 (d, J = 7.6 Hz, 1H).
  • 12B: (12 mg, 11% yield) Rt=7.25 minutes (analytical method); MS (LCMS) 411.3 (M+H+; positive mode); 409.1 (M-H+; negative mode). 1H NMR (400 MHz, methanol-d4) delta ppm 7.52-7.45 (m, 2H), 7.25 (d, J = 8.4 Hz, 2H), 7.19 (d, J = 8.4 Hz, 2H), 7.05 (dd, J = 9.8, 8.6 Hz, 1H), 4.05 (d, J = 7.2 Hz, 1H), 3.98 (s, 2H), 3.91-3.84 (m, 4H), 3.76 (d, J = 12.4 Hz, 1H), 3.52 (d, J = 7.6 Hz, 1H).
  • Example 13 (1S,2S,3S,4R,5S)-5-[4-fluoro-3-(4-(oxetan-3-yloxy)-benzyl)-phenyl)-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (13A)
  • Figure imgb0048
  • To a solution of ((2S,3S)-2,3,4-tris-benzyloxy-5-{4-fluoro-3-[4-(oxetan-3-yloxy)-benzyl]-phenyl}-6,8-dioxa-bicyclo[3.2.1]oct-1-yl)-methanol I-13k (300 mg, 0.417 mmol) in a 4:1 solution of ethanol/tetrahydrofuran (10 mL) was added successively formic acid (333 microL, 8.34 mmol) and palladium black (266 mg, 2.50 mmol). The resulting mixture was stirred at room temperature. After 3 hours, additional formic acid (333 microL, 8.34 mmol) and palladium black (266 mg, 2.50 mmol) were added. After 5 hours, the reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The resulting crude residue was purified by flash chromatography over silica gel (eluting with a gradient of 0 to 15% methanol in dichloromethane) to afford 153.0 mg of a white solid (mixture of isomers). The mixture of isomers was purified by preparative HPLC.
  • HPLC preparative method: reverse phase C18 Gemini column, 5 micrometer 30x100mm, 40mL/minute flow rate, gradient of acetonitrile/0.1 % formic acid : water/0.1 % formic acid; 25 to 50% acetonitrile/0.1 % formic acid over 18 minutes; UV detection: 220 nm.
  • 13A: (23 mg, 12% yield) Rt = 7.9 minutes; the fractions containing the product were concentrated under reduced pressure.
  • 1H NMR (400 MHz, methanol-d4) delta ppm 3.52 (d, J=7.8 Hz, 1H), 3.57 (d, J=7.2 Hz, 1H), 3.60 - 3.68 (m, 2 H), 3.75 (d, J=8.2 Hz, 1H), 3.81 (d, J=12.5 Hz, 1H), 3.89 (s, 2 H), 4.12 (d, J=7.4 Hz, 1 H), 4.63 (dd, J=7.3, 4.8 Hz, 2 H), 4.95 (t, J=6.5 Hz, 2 H), 5.16 - 5.23 (m, 1 H), 6.63 (m, 2 H), 7.00 (dd, J=9.7, 8.5 Hz, 1 H), 7.10 (m, 2 H), 7.36 - 7.42 (m, 2 H).
  • Example 14 (1S,2S,3S,4R,5S)-5-{4-chloro-3-[4-(oxetan-3-yloxy)-benzyl]-phenyl}-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (14A)
  • Figure imgb0049
  • To a solution of intermediate ((2S,3S)-2,3,4-tris-benzyloxy-5-{4-chloro-3-[4-(oxetan-3-yloxy)-benzyl]-phenyl}-6,8-dioxa-bicyclo[3.2.1]oct-1-yl)-methanol I-14k (182 mg) in ethanol/tetrahydrofuran (14 mL, 4/1 volume) was added successively formic acid (190 microL, 20 equivalents) and palladium black (106 mg, 4 equivalents) and the resulting mixture was stirred at room temperature. After 2 hours an additional 1 mL of tetrahydrofuran was added and the resulting mixture was stirred at room temperature for a additional hour. At this point additional formic acid (190 microL, 20 equivalents) and palladium black (106 mg, 4 equivalents) were added and the mixture was allowed to stir for an additional hour at room temperature. The palladium was filtered and the crude mixture obtained after evaporation of solvent (containing a mixture of isomers) was purified by HPLC preparative.
  • HPLC preparative method: reverse phase C18 Xbridge column 5 micrometer 100 x 30 mm, flow rate 40 mL/minute, gradient of acetonitrile/0.1 % formic acid : water/0.1 %formic acid; 30 to 55% of acetonitrile/0.1 % formic acid over 11 minutes; UV detection: 220 nm.
  • 14A: (20 mg, 17% yield); Rt = 4.43 minutes; the fractions containing the product were concentrated under reduced pressure resulting in a white solid.
  • MS (LCMS) 465.3 (M+H+; positive mode); 509.2 (M+HCO2 -; negative mode). 1H NMR (400 MHz, methanol-d4) delta ppm 3.53 (d, J=8.0 Hz, 1 H), 3.58 (dd, J=7.4, 1.4 Hz, 1 H), 3.64 (t, J=8.2 Hz, 1 H), 3.67 (d, J=12.4 Hz, 1 H), 3.77 (dd, J=8.4, 1.4 Hz, 1 H), 3.83 (d, J=12.6 Hz, 1 H), 4.03 (s, 2 H), 4.14 (d, J=7.4 Hz, 1 H), 4.65 (m, 2 H), 4.97 (t, J=6.6 Hz, 2 H), 5.22 (m, 1 H), 6.65 (m, 2 H), 7.11 (m, 2 H), 7.34 (d, J=8.4 Hz, 1 H), 7.38 (dd, J=8.4, 2.2 Hz, 1 H), 7.45 (d, J=2.0 Hz, 1 H).
  • Example 15 Cocrystalization of (1S,2S,3S,4R,5S)-5-[4-chloro-3-[4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (Example 4A compound) with L-proline to give (15):
  • L-proline dissolved in water (approximately 480 mg/mL) was added to the example 4A compound (approximately 80 moles L-proline per mole (example 4A compound)). Volume was doubled with ethanol and solution was capped and stirred for approximately 12 hours. Volume was reduced by half by evaporation on the bench. Volume was doubled using ethanol and the volume of solution was again reduced by half using evaporation. Solid was recovered using centrifugal filtration.
  • Example 16 Cocrystalization of (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicvclo[3.2.1]octane-2,3,4-triol (Example 4A compound) with L-proline to give (16):
  • L-proline dissolved in water (approximately 480 mg/mL) was added to the example 4A compound (approximately 59 moles L-proline per mole example 4A compound). Volume was doubled with methanol and solution was clear. Volume was increased by 25% using acetone. Solution was capped and stirred for approximately 12 hours. Volume was reduced by approximately 60% through evaporation on the bench. Volume was doubled using methanol and remaining solvent was evaporated leaving solid white precipitate.
  • Example 17 Cocrystalization of (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6, 8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (Example 4A compound) with L-proline to give (17):
  • A solution of ethanol saturated with L-proline was added to the example 4A compound (approximately 2.2 moles L-proline per mole example 4A compound) in a glass vial. Clear solution was capped and stirred for approximately 72 hours. Volume was reduced by half by evaporation at room temperature. Precipitate was seen and vial was capped and stirred for approximately 12 hours. White solid was collected using centrifugal filtration.
  • Example 18 Cocrystalization of (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzy/)-1pheny/]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (Example 4A compound) with L-proline to give (18):
  • L-proline dissolved in water (330 mg/mL) was dripped into approximately 2 mL of the example 4A compound dissolved in isopropanol (98 mg/mL) until solution became cloudy. After 15-20 minutes precipitation was observed and suspension became thick. Approximately 8 mL of water was added and solution was capped and stirred overnight. White solid was collected using vacuum filtration and dried in a 50°C vac oven for approximately 2 hours.
  • Example 19 Cocrystalization of (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (Example 4A compound) with L-Pyroglutamic acid to give (19):
  • 153 microL of the compound (4A) in isopropyl alcohol (97.97 mg/mL) was pipetted into 500 microL of L-pyroglutamic acid in water (213.0 mg/mL). Solution was capped and stirred overnight. Approximately 5-10 mg more solid L-pyroglutamic acid was added. 100 microL of ethanol was added. Solution was capped and stirred overnight. Ethanol was added until total volume was adjusted to approximately 2 mL. Solution was uncapped and left in hood overnight. Approximately 10-30 mg more example 4A compound was added. Solution was capped and stirred for approximately 2 days. White precipitate was seen. Suspension was pipetted into a Co-star microcentrifuge tube equipped with a 0.45 microm nylon filter membrane insert. Solution was centrifuged until solid was separated from solution. Cocrystal (19) was recovered.
  • Example 20 Cocrystalization of (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicvclo[3.2.1]octane-2,3,4-triol (Example 4A compound) with L-pyroglutamic acid to give (20):
  • 4-5 mL of a 1:1 ethanol/water solution was saturated with L-pyroglutamic acid (412.1 mg/mL). 730 mg of the example 4A compound was added to 3.2 mL of the L-pyroglutamic acid solution. After approximately 1 minute, precipitation was seen. The solution was too thick to stir therefore 2 mL of a 1:1 ethanol/water solution was added. The solution was stirred overnight. The solid was collected using vacuum filtration on a 0.45 microm nylon filter membrane. The solid was dried in a 50 °C vacuum oven for approximately 2 hours. Approximately 960 mg of the cocrystal complex (20) was recovered. The stoichiometric ratio of Example 4A compound to L-pyroglutamic acid was determined using quantitative NMR to be 1:1.63. Excess L-pyroglutamic acid was removed by suspending the material in ethanol yielding 1:1 co-crystal (20).
  • Example 21 Cocrystalization of (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (Example 4A compound) with L-pyroglutamic acid to give (21):
  • 494 mg of the example 4A compound was dissolved in 1.5 mL solution of isopropanol and ethanol (4:1 respectively). 917.2 mg of L-pyroglutamic acid was dissolved in 3 mL water. Both solutions were heated to 40°C. 200 microL of L-pyroglutamic acid solution was added to example 4A compound solution every minute until all solution was transferred (both solutions capped unless solution was being transferred). Vial with L-pyroglutamic acid solution was washed with 200 microL ethanol and solution was transferred to example 4A compound solution. Solution was stirred for 5 minutes and then heat was turned off (solution cooled at approximately 1 degree Celsius every 3 minutes). At 30°C, solution was placed on ambient temperature stirrer and stirred at 20°C for 20 minutes. Solution was clear. Approximately 2 mL of dry seeds were added. Suspension became thick over the next 2 hours. Solution was stirred overnight. Solid was recovered using vacuum filtration on a Pyrex 2 mL 10-15M sintered glass funnel filter. Solid was dried for 24 hours in a 50°C vacuum oven.
  • Example 22 Cocrystal of (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (Example 4A compound) and L-proline and cocrystal of (1S,2S,3S,4R,5S)-5-(4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (Example 4A compound) and L-pyroglutamic acid:
  • Powder X-ray Diffraction Analysis: The powder X-ray diffraction patterns of the cocrystal of example 4A compound with L-proline and the cocrystal of example 4A compound with L-pyroglutamic acid were carried out on a Bruker D5000 diffractometer using copper radiation (wavelength: 1.54056Å). The tube voltage and amperage were set to 40 kV and 40mA, respectively. The divergence and scattering slits were set at 1 mm, and the receiving slit was set at 0.6 mm. Diffracted radiation was detected by a Kevex PSI detector. A theta-two theta continuous scan at 2.4 ° per minute (1 second per 0.04° step) from 3.0 to 40 ° 2θ was used. An alumina standard was analyzed to check the instrument alignment. Data were collected and analyzed using Bruker axis software Version 7.0. Samples were prepared by placing them in a quartz holder. It should be noted that Bruker Instruments purchased Siemens; thus, Bruker D5000 instrument is essentially the same as a Siemens D5000. Eva Application 13.0.0.3 software was used to visualize and evaluate PXRD spectra. PXRD data files (.raw) were not processed prior to peak searching. Generally, a Threshold value of 2 and a Width value of 0.3 were used to make preliminary peak assignments. The output of automated assignments was visually checked to ensure validity and adjustments manually made if necessary.
  • To perform an X-ray diffraction measurement on a Bragg-Brentano instrument like the Bruker system used for measurements reported herein, the sample is typically placed into a holder which has a cavity. The sample powder is pressed by a glass slide or equivalent to ensure a random surface and proper sample height. The sample holder is then placed into the instrument. The incident X-ray beam is directed at the sample, initially at a small angle relative to the plane of the holder, and then moved through an arc that continuously increases the angle between the incident beam and the plane of the holder. Measurement differences associated with such X-ray powder analyses result from a variety of factors including: (a) errors in sample preparation (e.g., sample height), (b) instrument errors (e.g. flat sample errors), (c) calibration errors, (d) operator errors (including those errors present when determining the peak locations), and (e) the nature of the material (e.g. preferred orientation and transparency errors). Calibration errors and sample height errors often result in a shift of all the peaks in the same direction. Small differences in sample height when using a flat holder will lead to large displacements in XRPD peak positions. A systematic study showed that, using a Shimadzu XRD-6000 in the typical Bragg-Brentano configuration, sample height difference of 1 mm lead to peak shifts as high as 1 °2θ (Chen et al.; J Pharmaceutical and Biomedical Analysis, 2001; 26,63). These shifts can be identified from the X-ray Diffractogram and can be eliminated by compensating for the shift (applying a systematic correction factor to all peak position values) or recalibrating the instrument. As mentioned above, it is possible to rectify measurements from the various machines by applying a systematic correction factor to bring the peak positions into agreement. In general, this correction factor will bring the measured peak positions from the Bruker into agreement with the expected peak positions and may be in the range of 0 to 0.2 ° 2θ.
  • The powder X-ray diffraction values are generally accurate to within ± 0.2 2-theta degrees, due to slight variations of instrument and test conditions.
  • Cocrystal of example 4A compound and L-proline from Example 18 characterized by the following powder x-ray diffraction pattern, provided in Figure 3, expressed in terms of the degree 2θ and relative intensities with a relative intensity of ≥ 2.7% measured on a Bruker D5000 diffractometer with CuKα radiation:
    Angle (Degree 2θ) Relative Intensity* (≥2.7%)
    4.6 5.1
    5.5 12.8
    7.6 40.2
    8.5 11.9
    10.3 9.1
    11.0 4.5
    12.1 22.3
    12.6 13.5
    14.4 13.1
    14.8 16.1
    15.3 2.7
    15.9 10.4
    16.5 3.0
    16.8 8.2
    17.0 16.6
    17.4 33.9
    18.1 2.9
    18.4 10.3
    18.9 16.8
    19.5 12.2
    20.3 100.0
    21.0 6.5
    22.0 5.5
    22.2 7.1
    22.6 11.5
    22.9 29.3
    23.5 4.5
    24.3 13.8
    24.8 14.2
    25.4 14.7
    2.5.7 23.2
    26.0 6.9
    26.8 5.9
    27.0 5.8
    27.5 21.2
    28.8 15.5
    29.4 6.5
    29.8 8.2
    30.2 53
    30.7 14.1
    31.7 5.2
    32.1 7.4
    32.5 7.7
    33.0 9.9
    33.3 7.5
    33.8 5.5
    34.4 5.8
    35.5 3.4
    35.8 4.0
    36.9 3.1
    37.4 2.9
    38.2 4.7
    38.3 6.0
    39.3 8.0
    *The relative intensities may change depending on the crystal size and morphology.
  • Characteristic 2θ peaks or combinations of cocrystal of example 4A compound and L-proline:
    Angle (Degree 2θ)
    7.6
    12.1
    20.3
    28.8
  • Cocrystal of example 4A compound and L-pyroglutamic acid from Example 20 characterized by the following powder x-ray diffraction pattern, provided in Figure 4, expressed in terms of the degree 2θ and relative intensities with a relative intensity of ≥ 2.7% measured on a Bruker D5000 diffractometer with CuKα radiation:
    Angle (Degree 2θ) Relative Intensity* (≥4.3%)
    6.4 31.0
    7.6 5.9
    11.8 4.3
    12.3 8.8
    12.7 11.0
    13.5 10.4
    14.2 31.3
    14.4 24.0
    15.2 13.9
    16.7 66.8
    17.4 57.1
    18.3 10.5
    18.7 53.3
    19.1 24.2
    19.3 32.0
    19.8 9.2
    20.3 75.6
    21.1 100.0
    22.5 9.2
    23.6 11.7
    24.3 18.7
    24.7 22.2
    25.0 14.2
    26.2 53.4
    27.2 4.6
    27.9 10.0
    28.3 26.3
    29.0 14.5
    29.5 31.3
    30.7 16.2
    31.5 5.4
    32.0 23.2
    33.0 9.9
    34.2 19.9
    35.2 5.4
    35.9 13.6
    37.3 8.3
    37.9 9.5
    38.4 6.1
    39.2 10.3
    *The relative intensities may change depending on the crystal size and morphology.
  • Characteristic 2θ peaks or combinations of cocrystal of example 4A compound and L-pyroglutamic acid:
    Angle (Degree 2θ)
    6.4
    16.7
    17.4
    21.1
  • Example 23 Cocrystal of (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (Example 4A compound) and L-proline and cocrystal of (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (Example 4A compound) and L-pyroglutamic acid: Differential Scanning Calorimetry Thermogram Analysis:
  • Thermograms were obtained on a TA Instruments Q1000 Differential Scanning Calorimeter (DSC). 1-2 mg of sample was placed in aluminum sample pans and then covered with a pierced lid. The energy was measured against an empty pan as the temperature increased from 25°C to 200-300°C at 10°C per minute. The onset temperature of the melting endotherm was reported as the melting temperature. The onset temperature of the melting endotherm is dependent on the rate of heating, the purity of the sample, size of crystal and sample, among other factors. Typically, the DSC results are accurate to within about ±2°C, preferably to within ±1.5°C.
  • Example 18 cocrystal of Example 4A compound and L-proline DSC results are shown in Figure 5.
  • Example 20 cocrystal of Example 4A compound and L-pyroglutamic acid DSC results are shown in Figure 6.
  • Example 24 Cocrystal of (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (Example 4A compound) and L-proline
  • Single Crystal X-Ray Analysis. A representative crystal using the filtrate from example 17 and concentrating via solw evaporation was surveyed and a 0.85 Å data set (maximum sin Θ/λ=0.60) was collected on a Bruker APEX diffractometer. Friedel pairs were collected in order to facilitate the determination of the absolute configuration. Atomic scattering factors were taken from the International Tables for Crystallography, Vol. C, pp. 219, 500, Kluwer Academic Publishers, 1992. All crystallographic calculations were facilitated by the SHELXTL system, Version 5.1, Bruker AXS, 1997. All diffractometer data were collected at room temperature. Pertinent crystal, data collection, and refinement are summarized in Table 24-1.
  • A trial structure was obtained by direct methods. This trial structure refined routinely except the unexpected water molecule and the conformational disorder with the L-proline. The L-proline was modeled with ~60/40 occupancy in the "half-chair" and "envelope" conformations A very similar disorder was observed in H. D. Flack, Acta Crystallogr., A39, 876, 1983.
  • The hydrogen atoms bonded to N1, 06 and 07 located by difference Fourier techniques and allowed to refine with distances restrained. The relevant hydrogen atoms bonded to 05 were located from Fourier techniques, but was deleted and placed in idealized location (HFIX 83). The relevant hydrogen atom bonded to 04 could not be found with Fourier techniques and was placed in an idealized location (HFIX 83). The hydrogen atoms on the water molecule could not be located and were left out of the solution. The hydrogen parameters were added to the structure factor calculations but were not refined. The shifts calculated in the final cycles of least squares refinement were all less than 0.1 of the corresponding standard deviations. The final R-index was 5.15%. A final difference Fourier revealed no missing or misplaced electron density.
  • The refined structure was plotted using the SHELXTL plotting package (Figure 7). The absolute configuration was determined by the method of Flack4. Coordinates, anisotropic temperature factors, distances and angles are available as supplementary material (Tables 24-2 to 24-5). Table 24-1 Crystal data and structure refinement for Example 24
    Empirical formula C22 H25 Cl O7, C5 H9 N O2, H2O
    Formula weight 570.02
    Temperature 298(2) K
    Wavelength 1.54178 Å
    Crystal system Monoclinic
    Space group C2
    Unit cell dimensions a = 32.8399(16) Å α= 90°.
    b = 7.2457(4) Å β= 101.268(5)°.
    c = 11.8023(6) Å γ = 90°.
    Volume 2754.2(2) A3
    Z 4
    Density (calculated) 1.375 Mg/m3
    Absorption coefficient 1.729 mm-1
    F(000) 1208
    Crystal size 0.08 x 0.16 x 0.92 mm3
    Theta range for data collection 2.74 to 65.58°.
    Index ranges -38<=h<=37, -8<=k<=6, -13<=l<=13
    Reflections collected 6261
    Independent reflections 2922 [R(int) = 0.0526]
    Completeness to theta = 65.58° 74.9 %
    Absorption correction None
    Refinement method Full-matrix least-squares on F2
    Data / restraints / parameters 2922/5/380
    Goodness-of-fit on F2 0.953
    Final R indices [I>2sigma(I)] R1 = 0.0515, wR2 = 0.1304
    R indices (all data) R1 = 0.0581, wR2 = 0.1334
    Absolute structure parameter 0.02(3)
    Extinction coefficient 0.0027(2)
    Largest diff. peak and hole 0.252 and -0.210 e.Å-3
    Table 24-2. Atomic coordinates (x 104) and equivalent isotropic displacement parameters (Å2x 103) for Example 24. U(eq) is defined as one third of the trace of the orthogonalized Uij tensor.
    x y z U(eq)
    Cl(01) 7251(1) 14669(2) 1220(1) 73(1)
    N(1) 5586(1) 4836(6) 1285(3) 52(1)
    O(1) 6821(1) 9986(6) 5726(3) 66(1)
    C(1) 6691 (2) 13136(9) 6328(5) 78(2)
    O(2) 6311(1) 7637(5) -1948(2) 50(1)
    C(2) 6929(2) 11374(9) 6600(4) 66(1)
    O(3) 5936(1) 10106(4) -2723(2) 46(1)
    C(3) 6973(1) 10182(7) 4725(4) 55(1)
    O(4) 5415(2) 10596(7) -4969(4) 89(1)
    C(4) 7278(2) 11410(8) 4564(4) 62(1)
    C(5) 7411(2) 11447(8) 3506(4) 59(1)
    O(5) 5145(1) 6652(5) -3818(3) 66(1)
    C(6) 7237(1) 10299(7) 2618(4) 53(1)
    O(6) 5092(1) 7027(5) -1397(3) 63(1)
    O(7) 5878(1) 7968(5) 33(3) 54(1)
    C(7) 6932(2) 9079(7) 2793(4) 59(1)
    O(8) 5009(1) 2361(6) 1421 (4) 78(1)
    C(8) 6804(2) 8992(8) 3838(5) 59(1)
    O(9) 5409(1) -82(6) 1265(4) 76(1)
    C(9) 7365(1) 10429(8) 1443(4) 59(1)
    C(10) 7014(1) 11175(7) 533(4) 51(1)
    C(11) 6926(1) 13040(7) 382(4) 52(1)
    C(12) 6592(2) 13693(7) -395(4) 58(1)
    C(13) 6331(1) 12460(7) -1057(3) 52(1)
    C(14) 6401(1) 10583(6) -950(3) 44(1)
    C(15) 6744(1) 9959(7) -151 (3) 49(1)
    C(16) 6104(1) 9260(6) -1659(4) 47(1)
    C(17) 6125(1) 7179(7) -3112(4) 51(1)
    C(18) 5775(1) 8575(6) -3448(4) 49(1)
    C(19) 5720(2) 9220(7) -4695(4) 59(1)
    C(20) 5363(1) 8012(7) -3103(4) 51(1)
    C(21) 5455(1) 7344(7) -1843(4) 50(1)
    C(22) 5739(1) 8727(6) -1085(4) 46(1)
    C(23) 5335(2) 1609(8) 1299(4) 62(1)
    C(24) 5698(2) 2825(8) 1186(5) 63(1)
    C(25) 6072(2) 2583(12) 2141(9) 105(3)
    C(26) 5826(2) 5594(10) 2391 (5) 74(2)
    C(35A) 6000(5) 4000(30) 3036(13) 126(7)
    C(35B) 6229(4) 4430(20) 2538(15) 71(5)
    O(99A) 5382(2) 3257(7) 6727(5) 101(2)
    Table 24-3. Bond lengths [Å] and angles [°] for Example 24
    Cl(01)-C(11) 1.761 (4)
    N(1)-C(26) 1.492(7)
    N(1)-C(24) 1.513(7)
    N(1)-H(98A) 0.977(18)
    N(1)-H(98B) 1.00(2)
    O(1)-C(3) 1.377(5)
    O(1)-C(2) 1.434(7)
    C(1)-C(2) 1.499(8)
    C(1)-H(03C) 0.9600
    C(1)-H(03D) 0.9600
    C(1)-H(03E) 0.9600
    O(2)-C(17) 1.430(5)
    O(2)-C(16) 1.434(5)
    C(2)-H(03F) 0.9700
    C(2)-H(03G) 0.9700
    O(3)-C(16) 1.409(5)
    O(3)-C(18) 1.437(5)
    C(3)-C(4) 1.379(7)
    C(3)-C(8) 1.386(7)
    O(4)-C(19) 1.405(7)
    O(4)-H(4A) 0.8200
    C(4)-C(5) 1.401(7)
    C(4)-H(025) 0.9300
    C(5)-C(6) 1.371(7)
    C(5)-H(2) 0.9300
    O(5)-C(20) 1.400(6)
    O(5)-H(5) 0.8200
    C(6)-C(7) 1.383(7)
    C(6)-C(9) 1.528(6)
    O(6)-C(21) 1.413(5)
    O(6)-H(99A) 0.95(2)
    O(7)-C(22) 1.420(5)
    O(7)-H(99B) 0.93(2)
    C(7)-C(8) 1.380(7)
    C(7)-H(026) 0.9300
    O(8)-C(23) 1.235(7)
    C(8)-H(033) 0.9300
    O(9)-C(23) 1.251 (7)
    C(9)-C(10) 1.513(7)
    C(9)-H(02A) 0.9700
    C(9)-H(02B) 0.9700
    C(10)-C(11) 1.386(7)
    C(10)-C(15) 1.392(7)
    C(11)-C(12) 1.369(7)
    C(12)-C(13) 1.373(7)
    C(12)-H(027) 0.9300
    C(13)-C(14) 1.381(7)
    C(13)-H(021) 0.9300
    C(14)-C(15) 1.394(6)
    C(14)-C(16) 1.501(6)
    C(15)-H(030) 0.9300
    C(16)-C(22) 1.536(6)
    C(17)-C(18) 1.526(6)
    C(17)-H(02C) 0.9700
    C(17)-H(02D) 0.9700
    C(18)-C(19) 1.520(6)
    C(18)-C(20) 1.540(6)
    C(19)-H(03H) 0.9700
    C(19)-H(03I) 0.9700
    C(20)-C(21) 1.537(6)
    C(20)-H(4) 0.9800
    C(21)-C(22) 1.533(6)
    C(21)-H(015) 0.9800
    C(22)-H(013) 0.9800
    C(23)-C(24) 1.510(7)
    C(24)-C(25) 1.506(10)
    C(24)-H(029) 0.9800
    C(25)-C(35B) 1.479(17)
    C(25)-C(35A) 1.52(2)
    C(25)-H(34A) 0.9700
    C(25)-H(34B) 0.9700
    C(26)-C(35A) 1.440(19)
    C(26)-C(35B) 1.548(14)
    C(26)-H(03A) 0.9700
    C(26)-H(03B) 0.9700
    C(35A)-H(35A) 0.9700
    C(35A)-H(35B) 0.9700
    C(35B)-H(35C) 0.9700
    C(35B)-H(35D) 0.9700
    C(26)-N(1)-C(24) 109.1 (4)
    C(26)-N(1)-H(98A) 107(2)
    C(24)-N(1)-H(98A) 109(3)
    C(26)-N(1)-H(98B) 97(3)
    C(24)-N(1)-H(98B) 119(3)
    H(98A)-N(1)-H(98B) 114(4)
    C(3)-O(1)-C(2) 117.7(4)
    C(2)-C(1)-H(03C) 109.5
    C(2)-C(1)-H(03D) 109.5
    H(03C)-C(1)-H(03D) 109.5
    C(2)-C(1)-H(03E) 109.5
    H(03C)-C(1)-H(03E) 109.5
    H(03D)-C(1)-H(03E) 109.5
    C(17)-O(2)-C(16) 106.6(3)
    O(1)-C(2)-C(1) 113.4(5)
    O(1)-C(2)-H(03F) 108.9
    C(1)-C(2)-H(03F) 108.9
    O(1)-C(2)-H(03G) 108.9
    C(1)-C(2)-H(03G) 108.9
    H(03F)-C(2)-H(03G) 107.7
    C(16)-O(3)-C(18) 103.3(3)
    O(1)-C(3)-C(4) 125.4(4)
    O(1)-C(3)-C(8) 115.1(4)
    C(4)-C(3)-C(8) 119.4(4)
    C(19)-O(4)-H(4A) 109.5
    C(3)-C(4)-C(5) 119.7(5)
    C(3)-C(4)-H(025) 120.2
    C(5)-C(4)-H(025) 120.2
    C(6)-C(5)-C(4) 121.0(5)
    C(6)-C(5)-H(2) 119.5
    C(4)-C(5)-H(2) 119.5
    C(20)-O(5)-H(5) 109.5
    C(5)-C(6)-C(7) 118.6(4)
    C(5)-C(6)-C(9) 120.7(4)
    C(7)-C(6)-C(9) 120.7(4)
    C(21)-O(6)-H(99A) 105(3)
    C(22)-O(7)-H(99B) 108(3)
    C(8)-C(7)-C(6) 121.2(4)
    C(8)-C(7)-H(026) 119.4
    C(6)-C(7)-H(026) 119.4
    C(7)-C(8)-C(3) 120.0(4)
    C(7)-C(8)-H(033) 120.0
    C(3)-C(8)-H(033) 120.0
    C(10)-C(9)-C(6) 111.2(3)
    C(10)-C(9)-H(02A) 109.4
    C(6)-C(9)-H(02A) 109.4
    C(10)-C(9)-H(02B) 109.4
    C(6)-C(9)-H(02B) 109.4
    H(02A)-C(9)-H(02B) 108.0
    C(11)-C(10)-C(15) 116.7(4)
    C(11)-C(10)-C(9) 123.4(4)
    C(15)-C(10)-C(9) 119.8(4)
    C(12)-C(11)-C(10) 122.9(4)
    C(12)-C(11)-Cl(01) 117.6(4)
    C(10)-C(11)-Cl(01) 119.5(4)
    C(11)-C(12)-C(13) 119.1(5)
    C(11)-C(12)-H(027) 120.5
    C(13)-C(12)-H(027) 120.5
    C(12)-C(13)-C(14) 121.0(4)
    C(12)-C(13)-H(021) 119.5
    C(14)-C(13)-H(021) 119.5
    C(13)-C(14)-C(15) 118.6(4)
    C(13)-C(14)-C(16) 119.9(4)
    C(15)-C(14)-C(16) 121.4(4)
    C(10)-C(15)-C(14) 121.8(5)
    C(10)-C(15)-H(030) 119.1
    C(14)-C(15)-H(030) 119.1
    O(3)-C(16)-O(2) 105.4(3)
    O(3)-C(16)-C(14) 108.5(4)
    O(2)-C(16)-C(14) 111.6(3)
    O(3)-C(16)-C(22) 107.4(3)
    O(2)-C(16)-C(22) 110.3(4)
    C(14)-C(16)-C(22) 113.2(3)
    O(2)-C(17)-C(18) 104.8(3)
    0(2)-C(17)-H(02C) 110.8
    C(18)-C(17)-H(02C) 110.8
    O(2)-C(17)-H(02D) 110.8
    C(18)-C(17)-H(02D) 110.8
    H(02C)-C(17)-H(02D) 108.9
    O(3)-C(18)-C(19) 107.4(4)
    O(3)-C(18)-C(17) 100.7(3)
    C(19)-C(18)-C(17) 113.4(4)
    O(3)-C(18)-C(20) 106.7(3)
    C(19)-C(18)-C(20) 113.1(4)
    C(17)-C(18)-C(20) 114.3(4)
    O(4)-C(19)-C(18) 112.7(4)
    O(4)-C(19)-H(03H) 109.0
    C(18)-C(19)-H(03H) 109.0
    O(4)-C(19)-H(03I) 109.0
    C(18)-C(19)-H(03I) 109.0
    H(03H)-C(19)-H(031) 107.8
    O(5)-C(20)-C(21) 110.0(4)
    O(5)-C(20)-C(18) 113.5(4)
    C(21)-C(20)-C(18) 108.9(3)
    O(5)-C(20)-H(4) 108.1
    C(21)-C(20)-H(4) 108.1
    C(18)-C(20)-H(4) 108.1
    O(6)-C(21)-C(22) 110.6(3)
    O(6)-C(21)-C(20) 113.1(4)
    C(22)-C(21)-C(20) 109.9(4)
    O(6)-C(21)-H(015) 107.7
    C(22)-C(21)-H(015) 107.7
    C(20)-C(21)-H(015) 107.7
    O(7)-C(22)-C(21) 109.7(4)
    O(7)-C(22)-C(16) 111.8(3)
    C(21)-C(22)-C(16) 110.3(3)
    O(7)-C(22)-H(013) 108.3
    C(21)-C(22)-H(013) 108.3
    C(16)-C(22)-H(013) 108.3
    O(8)-C(23)-O(9) 127.9(5)
    O(8)-C(23)-C(24) 118.1(5)
    O(9)-C(23)-C(24) 114.1(5)
    C(25)-C(24)-C(23) 114.1(5)
    C(25)-C(24)-N(1) 103.3(5)
    C(23)-C(24)-N(1) 110.3(4)
    C(25)-C(24)-H(029) 109.6
    C(23)-C(24)-H(029) 109.6
    N(1)-C(24)-H(029) 109.6
    C(35B)-C(25)-C(24) 108.3(8)
    C(35B)-C(25)-C(35A) 42.4(8)
    C(24)-C(25)-C(35A) 103.1(8)
    C(35B)-C(25)-H(34A) 70.3
    C(24)-C(25)-H(34A) 111.2
    C(35A)-C(25)-H(34A) 111.2
    C(35B)-C(25)-H(34B) 137.2
    C(24)-C(25)-H(34B) 111.2
    C(35A)-C(25)-H(34B) 111.1
    H(34A)-C(25)-H(34B) 109.1
    C(35A)-C(26)-N(1) 104.8(9)
    C(35A)-C(26)-C(35B) 42.5(9)
    N(1)-C(26)-C(35B) 101.1(7)
    C(35A)-C(26)-H(03A) 110.8
    N(1)-C(26)-H(03A) 110.8
    C(35B)-C(26)-H(03A) 73.1
    C(35A)-C(26)-H(03B) 110.8
    N(1)-C(26)-H(03B) 110.8
    C(35B)-C(26)-H(03B) 144.0
    H(03A)-C(26)-H(03B) 108.9
    C(26)-C(35A)-C(25) 105.8(10)
    C(26)-C(35A)-H(35A) 110.6
    C(25)-C(35A)-H(35A) 110.6
    C(26)-C(35A)-H(35B) 110.6
    C(25)-C(35A)-H(35B) 110.6
    H(35A)-C(35A)-H(35B) 108.7
    C(25)-C(35B)-C(26) 102.7(8)
    C(25)-C(35B)-H(35C) 111.2
    C(26)-C(35B)-H(35C) 111.2
    C(25)-C(35B)-H(35D) 111.2
    C(26)-C(35B)-H(35D) 111.2
    H(35C)-C(35B)-H(35D) 109.1
    Table 24-4. Anisotropic displacement parameters (Å2x 103) for Example 24. The anisotropic displacement factor exponent takes the form: -2π2[ h2 a*2U11 + ... + 2 h k a* b* U12]
    U11 U22 U33 U23 U13 U12
    CI(01) 77(1) 69(1) 69(1) -11(1) 1(1) -27(1)
    N(1) 56(2) 40(2) 60(2) 0(2) 12(2) 0(2)
    O(1) 70(2) 70(3) 62(2) 2(2) 23(2) -3(2)
    C(1) 80(3) 76(4) 77(3) -2(3) 13(3) 8(3)
    O(2) 47(1) 52(2) 49(1) -1(1) 3(1) 12(1)
    C(2) 63(3) 84(4) 52(2) 1(2) 13(2) 7(3)
    O(3) 46(1) 43(2) 45(1) 2(1) 3(1) -1(1)
    C(3) 48(2) 59(3) 57(2) 5(2) 12(2) 8(2)
    O(4) 111(3) 83(3) 62(2) 8(2) -10(2) 23(3)
    C(4) 59(3) 72(4) 53(2) -10(2) 10(2) -12(2)
    C(5) 54(2) 66(3) 55(2) -6(2) 4(2) -11(2)
    O(5) 69(2) 59(2) 59(2) 2(2) -12(2) -8(2)
    C(6) 41(2) 55(3) 59(2) -2(2) 4(2) 4(2)
    O(6) 58(2) 47(2) 87(2) -6(2) 25(2) -9(1)
    O(7) 62(2) 49(2) 51(2) 2(1) 12(1) -1(1)
    C(7) 54(2) 59(3) 58(2) -11(2) -4(2) -2(2)
    O(8) 63(2) 63(3) 116(3) -22(2) 36(2) -12(2)
    C(8) 52(2) 57(3) 69(3) -1(2) 12(2) -7(2)
    O(9) 90(2) 52(3) 98(2) -8(2) 44(2) -7(2)
    C(9) 45(2) 72(4) 58(2) -12(2) 5(2) 1(2)
    C(10) 45(2) 61(3) 46(2) -4(2) 11(2) -3(2)
    C(11) 57(2) 48(3) 50(2) -8(2) 11(2) -16(2)
    C(12) 75(3) 43(3) 57(2) 2(2) 12(2) -1(2)
    C(13) 50(2) 54(3) 49(2) 7(2) 4(2) -2(2)
    C(14) 48(2) 44(2) 41(2) 1(2) 10(2) 0(2)
    C(15) 47(2) 54(3) 45(2) -5(2) 8(2) 5(2)
    C(16) 45(2) 49(3) 47(2) -1(2) 8(2) 3(2)
    C(17) 54(2) 47(3) 52(2) -10(2) 8(2) -2(2)
    C(18) 52(2) 44(3) 47(2) -6(2) 1(2) 0(2)
    C(19) 63(3) 60(3) 50(2) -7(2) 1(2) -3(2)
    C(20) 45(2) 45(3) 56(2) -3(2) -3(2) 4(2)
    C(21) 51(2) 37(2) 60(2) -1(2) 11(2) 3(2)
    C(22) 48(2) 40(2) 50(2) -2(2) 10(2) 5(2)
    C(23) 79(3) 46(3) 65(3) -12(2) 24(2) -3(2)
    C(24) 63(3) 49(3) 84(3) 3(2) 30(2) 4(2)
    C(25) 70(4) 63(5) 176(8) 13(5) 8(4) 14(3)
    C(26) 68(3) 79(4) 68(3) -3(3) -3(3) 8(3)
    C(35A) 104(11) 178(19) 79(8) 29(10) -26(8) -30(11)
    C(35B) 43(6) 73(10) 90(9) -3(7) -1(6) 4(6)
    O(99A) 130(4) 76(4) 95(3) 14(2) 18(3) 9(3)
    Table 24-5. Hydrogen coordinates (x 104) and isotropic displacement parameters (A2x 103) for Example 24
    x y z U(eq)
    H(98A) 5291(6) 4930(60) 1320(30) 33(9)
    H(98B) 5680(13) 5750(50) 760(30) 51(12)
    H(03C) 6764 13698 5659 117
    H(03D) 6757 13962 6973 117
    H(03E) 6398 12877 6176 117
    H(03F) 7223 11637 6698 80
    H(03G) 6878 10893 7327 80
    H(4A) 5481 11497 -4553 134
    H(025) 7394 12210 5155 74
    H(2) 7619 12262 3404 71
    H(5) 5097 7017 -4489 98
    H(99A) 4974(16) 8220(40) -1350(50) 64(15)
    H(99B) 5744(13) 8610(60) 540(30) 54(13)
    H(026) 6810 8302 2195 71
    H(033) 6604 8136 3947 71
    H(02A) 7604 11232 1504 71
    H(02B) 7443 9215 1214 71
    H(027) 6544 14955 -474 70
    H(021) 6103 12894 -1584 62
    H(030) 6793 8697 -74 59
    H(02C) 6017 5929 -3159 61
    H(02D) 6325 7285 -3614 61
    H(03H) 5982 9696 -4830 71
    H(03I) 5643 8173 -5204 71
    H(4) 5187 9111 -3149 61
    H(015) 5605 6171 -1818 59
    H(013) 5578 9843 -1009 55
    H(029) 5776 2606 438 76
    H(34A) 6327 2839 1870 126
    H(34B) 6084 1342 2453 126
    H(03A) 6044 6414 2248 89
    H(03B) 5645 6267 2805 89
    H(35A) 5809 3518 3496 152
    H(35B) 6259 4310 3548 152
    H(35C) 6415 4913 2066 85
    H(35D) 6372 4404 3339 85
  • Example 25 Cocrystal From Example 20 of (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6.8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (Example 4A compound) and L-pyroglutamic acid:
  • Single Crystal X-Ray Analysis. A representative crystal of a sample from Example 20 was surveyed and a 0.90 Å data set (maximum sin Θ/λ=0.56) was collected on a Bruker APEX diffractometer. Friedel pairs were collected in order to facilitate the determination of the absolute configuration. Stereochemistry determined from the flack parameter and also from the known chirality of the coformer (L-pyroglutamic acid). Atomic scattering factors were taken from the International Tables for Crystallography, Vol. C, pp. 219, 500, Kluwer Academic Publishers, 1992. All crystallographic calculations were facilitated by the SHELXTL Version 5.1, Bruker AXS, 1997 system. All diffractometer data were collected at room temperature. Pertinent crystal, data collection, and refinement are summarized in Table 25-1.
  • A trial structure was obtained by direct methods. This trial structure refined routinely, except for a low residual peak that was refined as 0.1 stoichiometric water. The stoichiometry of water was found by first deleting a hydroxyl group on the molecule, refining and measuring the resulting q peak; then comparing this peak to the residual peak from the water molecule. Using this method, a ratio of 1 to 0.1 (molecule to water) was estimated. Additionally, removing the water molecule from the solution and searching for void spaces in the crystal with Material Studio, Platon, and Mercury revealed a plausible volume of 33 cubic angstroms for a water molecule (water typically has about 40 cubic angstroms of space). The hydrogen atoms on nitrogen and oxygen were located by difference Fourier techniques and allowed to refine freely with no restraints. A few of the protons bonded to heteroatoms (H97a, H97b, H97c, and H97c) exhibited somewhat short bond lengths (~0.8 angstroms found vs. ~0.96 expected), yet the distances were left unrestrained. The hydrogen atoms on 099 (water) were not found from the difference map and left out of the structure solution. The hydrogen parameters were added to the structure factor calculations but were not refined. The shifts calculated in the final cycles of least squares refinement were all less than 0.2 of the corresponding standard deviations. The final R-index was 3.58%. A final difference Fourier revealed no missing or misplaced electron density. Of the residuals left, one is in a reasonable position for a proton bonded to 039 (carboxylic acid). This residual could be an additional occupancy position for the H98a proton (proton bonded to 039), but was not refined as such
  • The refined structure was plotted using the SHELXTL plotting package (Figure 8). The absolute configuration was determined by the method of Flack (H. D. Flack, Acta Crystallogr., A39, 876, 1983). Coordinates, anisotropic temperature factors, distances and angles are available as supplementary material (Tables 25-2 through 25-5). Table 25-1. Crystal data and structure refinement for Example 25.
    Empirical formula C22 H25 Cl1 O7 * C5 H7 N1 O3 * 0.1 (H2O)
    Formula weight 567.79
    Temperature 570(2) K
    Wavelength 1.54178 Å
    Crystal system Orthorhombic
    Space group P2(1)2(1)2(1)
    Unit cell dimensions a = 7.4907(10) Å α= 90°.
    b = 12.8626(15) Å β= 90°.
    c = 28.029(4) Å γ = 90°.
    Volume 2700.6(6) Å3
    Z 4
    Density (calculated) 1.396 Mg/m3
    Absorption coefficient 1.767 mm-1
    F(000) 1196
    Crystal size 0.03 x 0.2 x 0.2 mm3
    Theta range for data collection 3.15 to 59.28°.
    Index ranges -6<=h<=7, -13<=k<=14, -31 <=l<=29
    Reflections collected 9116
    Independent reflections 3759 [R(int) = 0.0275]
    Completeness to theta = 59.28° 96.5 %
    Absorption correction Empirical
    Refinement method Full-matrix least-squares on F2
    Data / restraints / parameters 3759 / 0 / 387
    Goodness-of-fit on F2 1.032
    Final R indices [I>2sigma(I)] R1 = 0.0358, wR2 = 0.0885
    R indices (all data) R1 = 0.0418, wR2 = 0.0920
    Absolute structure parameter 0.010(18)
    Extinction coefficient 0.00067(17)
    Largest diff. peak and hole 0.171 and -0.136 e.Å-3
    Table 25-2. Atomic coordinates (x 104) and equivalent isotropic displacement parameters (Å2x 103) for Example 25. U(eq) is defined as one third of the trace of the orthogonalized Uij tensor.
    x y z U(eq)
    C(1) 1385(7) 9812(3) 634(2) 89(1)
    C(2) 1302(6) 8776(3) 399(2) 81(1)
    O(3) 2546(3) 8104(1) 625(1) 55(1)
    C(4) 2553(4) 7080(2) 489(1) 43(1)
    C(5) 1555(5) 6676(2) 117(1) 54(1)
    C(6) 1669(4) 5630(2) 13(1) 50(1)
    C(7) 2745(4) 4966(2) 268(1) 42(1)
    C(8) 3723(4) 5385(2) 639(1) 50(1)
    C(9) 3643(4) 6423(2) 749(1) 49(1)
    C(10) 2839(5) 3807(2) 157(1) 49(1)
    C(11) 2248(4) 3159(2) 577(1) 40(1)
    C(12) 467(4) 3074(2) 708(1) 44(1)
    CI(13) -1197(1) 3652(1) 362(1) 59(1)
    C(14) -56(4) 2535(2) 1112(1) 46(1)
    C(15) 1219(4) 2055(2) 1391(1) 44(1)
    C(16) 3014(4) 2095(2) 1265(1) 36(1)
    C(17) 3502(4) 2652(2) 864(1) 38(1)
    C(18) 4335(3) 1493(2) 1560(1) 36(1)
    O(19) 3862(2) 420(1) 1544(1) 38(1)
    C(20) 5478(4) -95(2) 1690(1) 39(1)
    C(21) 5397(4) -1208(2) 1525(1) 50(1)
    O(22) 4012(4) -1782(2) 1739(1) 62(1)
    C(23) 5592(4) 18(2) 2233(1) 40(1)
    O(24) 7085(3) -534(2) 2419(1) 52(1)
    C(25) 5750(4) 1168(2) 2361(1) 43(1)
    O(26) 5536(3) 1267(2) 2861(1) 61(1)
    C(27) 4393(4) 1816(2) 2086(1) 39(1)
    O(28) 4750(3) 2904(2) 2136(1) 50(1)
    O(29) 6093(3) 1545(1) 1363(1) 44(1)
    C(30) 6903(4) 529(2) 1425(1) 47(1)
    O(31) 8289(3) 3646(2) 2167(1) 62(1)
    C(32) 8567(4) 4451(3) 1935(1) 50(1)
    C(33) 7407(5) 4857(3) 1544(1) 60(1)
    C(34) 8309(7) 5813(3) 1364(2) 100(2)
    C(35) 10008(5) 5962(3) 1666(1) 60(1)
    N(36) 9939(4) 5092(2) 1990(1) 62(1)
    C(37) 10097(5) 7035(3) 1889(1) 62(1)
    O(38) 10174(4) 7793(2) 1636(1) 94(1)
    O(39) 10136(4) 7059(2) 2353(1) 72(1)
    O(99) 470(40) 9300(20) 1968(10) 97(10)
    Table 25-3. Bond lengths [Å] and angles [°] for Example 25.
    C(1)-C(2) 1.486(5)
    C(2)-O(3) 1.419(4)
    O(3)-C(4) 1.371(3)
    C(4)-C(9) 1.382(4)
    C(4)-C(5) 1.385(4)
    C(5)-C(6) 1.379(4)
    C(6)-C(7) 1.375(4)
    C(7)-C(8) 1.381(4)
    C(7)-C(10) 1.525(4)
    C(8)-C(9) 1.371(4)
    C(10)-C(11) 1.509(4)
    C(11)-C(12) 1.388(4)
    C(11)-C(17) 1.398(4)
    C(12)-C(14) 1.382(4)
    C(12)-CI(13) 1.746(3)
    C(14)-C(15) 1.381(4)
    C(15)-C(16) 1.391(4)
    C(16)-C(17) 1.381(4)
    C(16)-C(18) 1.504(4)
    C(18)-O(19) 1.426(3)
    C(18)-O(29) 1.430(3)
    C(18)-C(27) 1.532(4)
    O(19)-C(20) 1.440(3)
    C(20)-C(21) 1.507(4)
    C(20)-C(23) 1.529(4)
    C(20)-C(30) 1.529(4)
    C(21)-O(22) 1.408(4)
    C(23)-O(24) 1.424(3)
    C(23)-C(25) 1.527(4)
    C(25)-O(26) 1.418(3)
    C(25)-C(27) 1.523(4)
    C(27)-O(28) 1.430(3)
    O(29)-C(30) 1.452(3)
    O(31)-C(32) 1.239(4)
    C(32)-N(36) 1.327(4)
    C(32)-C(33) 1.494(4)
    C(33)-C(34) 1.491(5)
    C(34)-C(35) 1.539(5)
    C(35)-N(36) 1.443(4)
    C(35)-C(37) 1.517(5)
    C(37)-O(38) 1.206(4)
    C(37)-O(39) 1.301(4)
    0(3)-C(2)-C(1) 108.8(3)
    C(4)-O(3)-C(2) 117.8(2)
    O(3)-C(4)-C(9) 116.4(3)
    O(3)-C(4)-C(5) 124.5(2)
    C(9)-C(4)-C(5) 119.1(3)
    C(6)-C(5)-C(4) 119.5(3)
    C(7)-C(6)-C(5) 122.1(3)
    C(6)-C(7)-C(8) 117.4(3)
    C(6)-C(7)-C(10) 121.9(3)
    C(8)-C(7)-C(10) 120.7(3)
    C(9)-C(8)-C(7) 121.8(3)
    C(8)-C(9)-C(4) 120.1(3)
    C(11)-C(10)-C(7) 111.6(2)
    C(12)-C(11)-C(17) 117.2(2)
    C(12)-C(11)-C(10) 122.1(3)
    C(17)-C(11)-C(10) 120.6(3)
    C(14)-C(12)-C(11) 121.9(3)
    C(14)-C(12)-CI(13) 117.8(2)
    C(11)-C(12)-CI(13) 120.4(2)
    C(15)-C(14)-C(12) 119.5(3)
    C(14)-C(15)-C(16) 120.5(3)
    C(17)-C(16)-C(15) 118.8(3)
    C(17)-C(16)-C(18) 122.7(2)
    C(15)-C(16)-C(18) 118.4(2)
    C(16)-C(17)-C(11) 122.2(3)
    O(19)-C(18)-O(29) 105.1(2)
    O(19)-C(18)-C(16) 108.5(2)
    O(29)-C(18)-C(16) 111.6(2)
    O(19)-C(18)-C(27) 107.5(2)
    O(29)-C(18)-C(27) 109.5(2)
    C(16)-C(18)-C(27) 114.1(2)
    C(18)-O(19)-C(20) 103.16(19)
    O(19)-C(20)-C(21) 108.4(2)
    O(19)-C(20)-C(23) 106.7(2)
    C(21)-C(20)-C(23) 113.5(2)
    O(19)-C(20)-C(30) 101.9(2)
    C(21)-C(20)-C(30) 112.1 (2)
    C(23)-C(20)-C(30) 113.3(2)
    O(22)-C(21)-C(20) 113.3(3)
    O(24)-C(23)-C(25) 109.6(2)
    O(24)-C(23)-C(20) 111.2(2)
    C(25)-C(23)-C(20) 109.3(2)
    O(26)-C(25)-C(27) 112.1(2)
    O(26)-C(25)-C(23) 108.1(2)
    C(27)-C(25)-C(23) 111.1(2)
    O(28)-C(27)-C(25) 111.2(2)
    O(28)-C(27)-C(18) 111.4(2)
    C(25)-C(27)-C(18) 110.9(2)
    C(18)-O(29)-C(30) 107.28(19)
    O(29)-C(30)-C(20) 103.8(2)
    O(31)-C(32)-N(36) 126.0(3)
    O(31)-C(32)-C(33) 125.4(3)
    N(36)-C(32)-C(33) 108.6(3)
    C(34)-C(33)-C(32) 105.9(3)
    C(33)-C(34)-C(35) 106.9(3)
    N(36)-C(35)-C(37) 116.5(3)
    N(36)-C(35)-C(34) 102.8(3)
    C(37)-C(35)-C(34) 112.1(3)
    C(32)-N(36)-C(35) 115.8(3)
    O(38)-C(37)-O(39) 124.4(4)
    O(38)-C(37)-C(35) 119.8(3)
    O(39)-C(37)-C(35) 115.8(3)
    Table 25-4. Anisotropic displacement parameters (Å2x 103) for Example 25. The anisotropic displacement factor exponent takes the form: -2π2[ h2 a*2U11 + ... + 2 h k a* b*U12]
    U11 U22 U33 U23 U13 U12
    C(1) 115(4) 49(2) 104(3) -20(2) -54(3) 31(2)
    C(2) 95(3) 52(2) 95(3) -12(2) -47(2) 23(2)
    O(3) 62(1) 36(1) 68(1) -3(1) -17(1) 6(1)
    C(4) 45(2) 35(2) 49(2) 1(1) 1(1) 0(1)
    C(5) 61(2) 43(2) 59(2) 4(2) -16(2) 9(2)
    C(6) 61(2) 43(2) 47(2) 2(1) -13(1) 0(2)
    C(7) 50(2) 36(2) 41(1) 6(1) 7(1) -2(1)
    C(8) 58(2) 44(2) 49(2) 9(1) -9(2) 7(2)
    C(9) 54(2) 43(2) 50(2) 2(1) -12(1) 3(1)
    C(10) 63(2) 40(2) 43(2) 6(1) 5(1) 6(1)
    C(11) 52(2) 27(1) 41(1) 0(1) 4(1) -2(1)
    C(12) 50(2) 29(2) 52(2) 0(1) -4(1) -4(1)
    CI(13) 59(1) 51(1) 66(1) 7(1) -16(1) 4(1)
    C(14) 40(2) 35(2) 64(2) 7(1) 4(1) -2(1)
    C(15) 44(2) 39(2) 50(2) 11(1) 7(1) -4(1)
    C(16) 41(2) 25(1) 42(1) 3(1) 2(1) -3(1)
    C(17) 36(2) 31(2) 45(2) 0(1) 5(1) -3(1)
    C(18) 36(2) 28(1) 43(1) 3(1) 4(1) -7(1)
    O(19) 40(1) 28(1) 45(1) 2(1) -5(1) -5(1)
    C(20) 40(2) 30(2) 47(2) 1(1) -6(1) 2(1)
    C(21) 57(2) 33(2) 60(2) -3(2) -12(2) 6(1)
    O(22) 77(2) 37(1) 73(2) 6(1) -21(1) -14(1)
    C(23) 42(2) 32(2) 47(2) 2(1) -8(1) -2(1)
    O(24) 58(1) 37(1) 62(1) 8(1) -18(1) -3(1)
    C(25) 52(2) 36(2) 42(2) 0(1) -7(1) -8(1)
    O(26) 100(2) 40(1) 43(1) -1(1) -9(1) -9(1)
    C(27) 45(2) 28(1) 43(1) -2(1) 0(1) -6(1)
    O(28) 55(2) 28(1) 69(1) -6(1) -7(1) -3(1)
    O(29) 36(1) 39(1) 56(1) 8(1) 7(1) -4(1)
    C(30) 48(2) 38(2) 56(2) 2(1) -1(1) 8(1)
    O(31) 62(2) 56(1) 70(1) 7(1) -9(1) -12(1)
    C(32) 48(2) 45(2) 57(2) -9(2) -3(2) 1(2)
    C(33) 57(2) 64(2) 58(2) -7(2) -12(2) 2(2)
    C(34) 135(4) 78(3) 87(3) 18(2) -58(3) -19(3)
    C(35) 55(2) 61(2) 64(2) 5(2) 7(2) -3(2)
    N(36) 53(2) 61(2) 72(2) 11(2) -22(2) -9(2)
    C(37) 49(2) 65(2) 72(2) 5(2) -1(2) 6(2)
    O(38) 116(2) 70(2) 95(2) 19(2) -2(2) -11 (2)
    O(39) 79(2) 69(2) 70(2) 2(1) 0(1) 3(1)
    O(99) 90(20) 110(20) 89(19) -60(18) -2(15) 29(17)
    Table 25-5. Hydrogen coordinates (x 104) and isotropic displacement parameters (Å2x 103) for Example 25.
    x y z U(eq)
    H(1A) 1080 9742 965 134
    H(1B) 557 10275 482 134
    H(1C) 2571 10087 606 134
    H(2A) 107 8492 427 97
    H(2B) 1587 8843 63 97
    H(5) 813 7106 -61 65
    H(6) 998 5365 -239 60
    H(8) 4455 4952 819 60
    H(9) 4323 6685 999 59
    H(10A) 2083 3655 -115 58
    H(10B) 4055 3623 73 58
    H(14) -1256 2497 1194 56
    H(15) 875 1701 1665 53
    H(17) 4703 2691 782 45
    H(21A) 5235 -1219 1181 60
    H(21 B) 6528 -1543 1595 60
    H(97D) 3240(80) -1700(40) 1620(20) 120(20)
    H(23) 4498 -261 2375 48
    H(97C) 6720(50) -1040(30) 2557(13) 78(14)
    H(25) 6950 1407 2275 52
    H(97B) 5570(50) 1910(30) 2942(14) 79(13)
    H(27) 3214 1680 2224 46
    H(97A) 5800(50) 2990(30) 2137(12) 60(12)
    H(30A) 7992 577 1611 57
    H(30B) 7175 214 1119 57
    H(33A) 7294 4347 1291 71
    H(33B) 6226 5021 1664 71
    H(34A) 7528 6411 1397 120
    H(34B) 8616 5735 1030 120
    H(35) 11047 5874 1457 72
    H(99) 10740(50) 4950(20) 2171(11) 49(10)
    H(98A) 9670(60) 6410(30) 2507(15) 95(14)
  • Example 26 Cocrystal of (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (Example 4A compound) and L-pyroglutamic acid: Solid state NMR:
  • Approximately 80 mg of cocrystal of (1 S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol (Example 4A compound) and L-pyroglutamic acid prepared using the process described in Scheme 2 was tightly packed into a 4 mm ZrO2 rotor. Spectra were collected at room temperature and pressure on a Bruker-Biospin 4 mm BL CPMAS probe positioned into a wide-bore Bruker-Biospin DSX 500 MHz (1H frequency) NMR spectrometer. The packed rotor was oriented at the magic angle and spun at 15.0 kHz. The 13C solid state spectrum was collected using a proton decoupled cross-polarization magic angle spinning experiment (CPMAS). The cross-polarization contact time was set to 2.0 ms. A proton decoupling field of approximately 85 kHz was applied. 1448 scans were collected with recycle delay of 14 seconds. The carbon spectrum was referenced using an external standard of crystalline adamantane, setting its upfield resonance to 29.5 ppm. The chemical shift data is dependent on the testing conditions (i.e. spinning speed and sample holder), reference material, and data processing parameters, among other factors. Typically, the ss-NMR results are accurate to within about ± 0.2 ppm.
  • Carbon chemical shifts observed.
    Characteristic peaks are starred.
    All peaks are (± 0.2 ppm)
    13C Chemical Shifts
    [ppm]a
    Intensityb
    16.5* 9.3
    2.31 4.8
    30.5 4.5
    42.5 4.9
    56.7 4.5
    61.0 6.2
    63.0 7.6
    69.4 8.0
    70.0 7.2
    74.7 8.4
    80.2 6.0
    85.3 11.4
    110.1 10.0
    110.4 7.9
    117.7 7.1
    127.9 5.7
    128.7 6.2
    130.0 6.9
    131.1* 12.0
    132.2 6.5
    134.4 0.8
    137.8 5.9
    140.3 5.3
    158.7* 7.9
    174.7 4.5
    181.5* 5.0
    (a) Referenced to external sample of solid phase adamantane at 29.5 ppm.
    (b) Defined as peak heights. Intensities can vary depending on the actual setup of the CPMAS experimental parameters and the thermal history of the sample. CPMAS intensities are not necessarily quantitative.
  • The SSNMR 13C CPMAS spectrum of Example 26 is shown in Figure 9. The peaks marked by asterisks in Figure 9 are spinning sidebands.
  • PHARMACOLOGICAL TESTING
  • The practice of the instant invention for the treatment of diseases modulated by the inhibition of SGLT2 can be evidenced by activity in at least one of the protocols described hereinbelow.
  • Biological Assays In-Vitro Assay
  • The SGLT2 functional assay was designed to detect the inhibition of methyl-alpha-D glucopyranoside (AMG - a non-metabolizable form of glucose) uptake via the SGLT2 transporter. The SGLT2 transporter recovers glucose from the proximal tubules of the kidney; its inhibition results in sugar wasted in the urine. The positive control compound, Phlorizin, is a known inhibitor of glucose uptake for SGLT2 and was used for comparing the high percent effect of SGLT2 inhibition of the test compounds.
  • CHO-FlpIn (Invitrogen, Carlsbad, CA) cells stably expressing human SGLT2 (pcDNA5/FRT) were plated in Iso-TC 96 well plates (Perkin Elmer, Waltham, MA) at a density of 100,000 cells/well in 100 microL of growth media (1:1 F-12/DMEM media (Gibco, Carlsbad, CA), 10% FBS (Sigma, St. Louis MO), 1X Pen/Strep (Gibco, Carlsbad, CA), 600 microg/mL Hygromycin (Invitrogen, Carlsbad, CA)). Prior to treating with test compound, confluent cells were serum starved for 2 hours at 37°C in 1:1 F-12/DMEM media, replacing with fresh F-12/DMEM media after 1 hour. Test compounds in dimethylsulfoxide (Sigma, St. Louis, MO) were diluted 100 fold in uptake buffer (140mM NaCl (Promega, Madison, WI), 2mM KCI (Teknova, Hollister, CA), 1mM CaCl2 (Teknova, Hollister, CA), 1mM MgCl2 (Teknova, Hollister, CA), and 10mM HEPES (Gibco, Carlsbad, CA) to cell plates pre-rinsed with uptake buffer. Cells were pre-incubated with test compound for 15 minutes prior to the addition of 50 microL AMG (40 nCi AMG [U-14C] (Perkin Elmer, Waltham, MA) in unlabelled AMG (Aldrich, St. Louis, MO)) per well yielding a final concentration of 11.33 microM AMG. Cell plates were then incubated for 3 hours at 37°C for AMG uptake. After incubation, cells were washed twice with ice cold wash buffer (uptake buffer containing 200 microM Phlorizin (Sigma), air dried and lysed in 30 microL of 200 mM NaOH and 1% SDS buffer on an orbital shaker. Microscint 40 (Perkin Elmer, Waltham, MA) was added to the lysed cells (giving a final volume of 200 microL) and mixed by orbital shaking for 30 minutes. Plates were stored in the dark overnight and quantitated in the 1540 Microbeta Trilux (Wallac, Waltham, MA) using a normalized protocol for 14C detection. The percent effect of test compounds to inhibit AMG uptake was calculated using the following calculation: % Effect = ZPE - T / ZPE - HPE x 100 %
    Figure imgb0050

    where "ZPE" is the corrected counts per minute (CCPM) in control wells containing 0.5% DMSO, T is the CCPM in wells containing test compound at various concentrations of the standard curve, and HPE is the high percent effect referring to the CCPM in control wells containing 10 microM Phlorizin. The IC50 values were calculated using a dose response equation and are summarized for the compounds tested in Table 3.
  • Abbreviations used in the in vitro testing description include:
    • SGLT2 type 2 sodium/glucose co-transporter
    • AMG methyl-α-D Glucopyranoside
    • DMEM Dulbecco's Modified Eagle's Medium
    • IC50 50% Inhibition Concentration
    • FBS Fetal Bovine Serum
    • DMSO Dimethylsulfoxide
    • SDS Sodium Dodecyl Sulfate
    • CHO-FlpIn Chinese Hamster Ovary cell containing the FRT site
    Table 3
    Test Compound Run No. hSGLT1 IC50 nM hSGLT2 IC50 nM
    1A 1 1080 1.55
    2 454 1.15
    3 327 0.779
    4 562 0.715
    5 262 0.654
    6 359 1.61
    2A 1 1240 0.827
    2 >1000 1.53
    3 >1000 0.942
    4 >1000 0.741
    5 679 1.58
    6 undetermined 1.05
    3A 1 543 0.479
    2 397 0.972
    3 550 1.39
    4 757 0.811
    5 523 0.602
    6 672 0.588
    7 380 1.35
    3B 1 >10000 41.6
    2 >10000 40.8
    3 >10000 27.9
    4 undetermined 62.2
    4A 1 1590 1.27
    2 1010 0.816
    3 1750 0.57
    4 >1000 0.922
    5 >1000 1.85
    6 2090 0.812
    7 1810 0.7
    8 2860 0.737
    9 2480 0.846
    10 2840 0.768
    4B 1 >1000 122
    2 >10000 66.8
    3 undetermined 81.7
    5A 1 >10000 4.5
    2 >1000 81.7
    3 5790 2.42
    4 undetermined 1.77
    5B 1 >10000 186
    6A 1 >10000 18.7
    2 >1000 9.99
    3 >1000 13.5
    4 >1000 13.4
    5 8930 5.71
    6 undetermined 7.67
    7A 1 >1000 10.6
    2 >10000 6.38
    3 >1000 5.88
    4 undetermined 6.11
    10A 1 >10,000 4.08
    2 >3330 33.4
    3 >3160 2.54
    10B 1 > 10, 000 127
    2 > 10, 000 103
    11A 1 >10,000 9.6
    2 >10,000 11.9
    3 >10,000 19.8
    4 > 10, 000 7.13
    12A 1 > 10, 000 11.1
    2 5780 7.41
    3 >10,000 8.85
    4 >10,000 0.802
    5 >10,000 10.7
    6 >10,000 14.1
    12B 1 >3160 32.3
    13A 1 >10,000 14.9
    2 >10,000 17.8
    14A 1 >10,000 2.28
    2 >10,000 3.12
    3 >10,000 2.39
    4 >10,000 2.87
    In-Vivo Assay
  • Examples 1A and 4A were tested in rats to assess inhibition of glucose transport via urinary glucose excretion. Male Sprague Dawley rats (~300 g) were singly housed in metabolic cages for urine collection. Rats had access to standard laboratory chow and water ad libitum. Rats (n=2 to 5/group) received vehicle or compound by oral gavage. Dosing solutions were 0.03 mg/mL, 0.3 mg/mL, 0.9 mg/mL, 3 mg/mL, 9 mg/mL and 18 mg/mL for the 0.1 mg/kg, 1 mg/kg, 3 mg/kg, 10 mg/kg, 30 mg/kg and 60 mg/kg doses respectively. Dosing volume was 1 mL/300 g of body weight for all doses. One group received a 10 mg/kg dose of Example 1A and others received 0.1, 1, 3, 10, 30 or 60 mg/kg dose of Example 4A. The vehicle was 20% v/v PEG400 and 24% v/v hydroxypropyl beta cyclodextrin; HPBCD. Following oral administration, urine was collected for 24 hours. Glucose concentration was measured in urine by UV absorbance spectrophotometry at 340 nm using a Roche Hitachi 917 spectrophotometer (Diamond Diagnostics, Holliston, MA). The total amount of glucose excreted in the urine was calculated as the product of urine concentration and urine volume using the formula below:
    • urinary glucose excreted (mg)/200g body weight = urinary glucose concentration (mg/dL) x urine volume (dL) x 200/rat body weight (g). Amounts of urinary glucose excreted (UGE) were obtained from rats for Example 1A and Example 4A by the method described above and are shown in Table 4. Blood (0.1 mL) was collected from the PK satellite group animals at 1, 2, 4, 7, 24 hours postdose to obtain plasma and analyzed by LC-MS/MS. Mean PK parameters at the various doses tested are shown in Table 4.
    Table 4
    Compound Dose (mg/kg) Mean UGE (mg/200g of body weight) ± SEM (n=5) Mean PK Parameters (n=2)
    Cmax (ng/mL) tmax (h) AUC(0-24) (ng*h/mL)
    Example 1A 10 2049 ± 382.2 1260 1.00 6630
    Example 4A 0.1 389.0 ± 62.54 43.4 1.50 188
    Example 4A 1 1519 ± 52.02 372 1.00 2000
    Example 4A 3 1937 ± 101.1 1320 1.00 7080
    Example 4A 10 2145 ± 132.3 3100 2.50 26400
    Example 4A 30 2554 ± 141.1 10500 1.00 10700
    Example 4A 60 2437 ± 116.7 25300 2.00 233000
    SEM: Standard error of the mean.
    Pharmacokinetic Testing in Rats
  • Examples 1A, 2A, 4A, 12A, and 14A were tested in rats to assess pharmacokinetic parameters including maximum concentration (Cmax), area under the plasma concentration time curve (AUC), clearance (CL), steady state volume of distribution (Vss), half life (t1/2), and bioavailability (F). Male Sprague Dawley rats (~ 300 g) were used. Rats received compound by intraveneous (IV) or oral gavage (PO) administration and the doses tested including vehicle to formulate dosing solutions are listed in Table 5.
  • Following IV or PO administration, 0.2 mL blood was sampled from the jugular vein at various timepoints (Table 5). Twenty microL aliquots of plasma samples and standards were subjected to protein precipitation with acetonitrile containing an internal standard. Samples were vortexed and centrifuged to obtain supernatant which was analyzed by LC-MS/MS. Analyst (Version 1.4.1) was used to measure peak areas and peak area ratios of analyte to internal standard were calculated. LC-MS/MS conditions are as follows: Mass Spectrometer + Source Type was Sciex API 4000 - Turbo Spray; HPLC pump was Shimadzu; Autosampler was CTC PAL Autosampler; Injection volume was 3.0 to 10 microL; A gradient was used with mobile phase A: 10 mM ammonium acetate and 1% Isopropyl alcohol in water; B: Acetonitrile; Flowrate 0.300 mL per minute (Column 2.0 x 30 mm 5 microm LUNA C18 column (phenomenex)). Detection mode was negative.
  • A calibration curve was constructed from the peak area ratios of the standards to the internal standard by applying a weighted linear (1/x or 1/x2) regression. The dynamic range of the standard curve was 5.00 ng/mL to 5000 ng/mL.
  • Pharmacokinetic parameters were determined from individual animal data using non-compartmental analysis in Watson (version 7.2). Concentrations below the limit of quantitation (BLQ) were recorded as 0 ng/mL for use in calculations.
    The following calculations were used:
    • AUC(0-τ) = Determined using the linear trapezoidal method
    • AUC(0-∞) = AUC(0-τ) plus extrapolated area determined by dividing plasma concentration at τ by the slope of the terminal log-linear phase
    • CL = Dose/AUC(0-∞)
    • Vdss = CL x MRT
    • Cmax = Recorded directly from plasma concentration time curve
    • Tmax = Recorded directly from plasma concentration time curve
    • t1/2 = In(0.5)/slope of the terminal log-linear phase
    • F% = AUC(0-∞)PO per dose/AUC(0-∞)IV per dose
    • C(0) = Extrapolated by linear regression from the apparent distribution phase following IV administration
    • MRT = AUMC (AUC(0-∞)/AUC(0-∞)
    Table 5
    Example Dose (mg/kg) Route /n Formulation Time points (h) Cmax (µg/mL) AUCinf (µg*h/mL) CL (mL/min/kg) Vss (Ukg) t1/2 (h) F (%)
    4A 2 IV / (n=2) DMSO/ PEG400/ 30%SBECD (10/30/60 v/v/v) 0 083. 0.25. 0.5. 1.2.4.6. 8. 20 - 8.48 4.04 1.1 4.1 -
    2 PO / (n=3) Tween 80/ 0.5% MC (0.1/99.9 v/v) 0.5, 1, 2, 4, 7, 20 0.772 5.65 - - 3.7 67
    5 PO / (n=5) 20% PEG / 24% HBCD 1, 4, 7, 24 1.19 16.8 - - - 79
    12A 2 IV / (n=2) DMSO/ PEG400/ 30%SBECD (5/10/85 v/v/v) 0.083, 0.25, 0.5, 1, 2, 4, 5, 6, 7, 20 - 2.20 15.9 3.68 3.90 -
    1A 2 IV / (n=2) DMA/ PG/ 50 mM Tris Base (5/10/85 v/v/v) 0.083, 0.25, 0.5, 1, 2, 4, 6, 8, 20 - 0.947 37.1 1.71 0.962 -
    10 PO / (n=2) PEG200/ 0.5%MC (5/ 95 v/v) 0.25, 0.5, 0.75, 1, 2, 4, 6, 8,20 1.65 2.68 - - 2.82 56.5
    14A 2 IV / (n=2) DMSO/ PEG400/ 30%SBECD (5/10/85 v/v/v) 0.25, 0.5, 1, 2, 4, 7, 20,22 - 1.06 31.7 1.69 1.36 -
    10 PO / (n=2) PEG200/ 0.5%MC (5/ 95 v/v) 0.5, 1, 2, 4, 7, 20, 22 0.551 2.29 - - 1.71 43.5
    2A 2 IV / (n=2) DMA/ PG/ 50 mM Tris Base (5/10/85 v/v/v) 0 083, 0.25, 0 5, 1, 2, 4, s, 8, 20 - 1.34 27.7 1.03 0.94 -
    3A 2 IV/ (n=2) DMA/ PG/ 50 mM Tris Base (5/10/85 v/v/v) 0.083, 0.25, 0.5, 1, 2, 4, 6, 8, 20 - 1.41 23.8 1.82 1.58 -
    - = Data not available or not applicable; DMSO = Dimethyl Sulfoxide; HBCD = Hydroxypropyl beta cyclodextrin; PEG = Polyetheylene Glycol; PG = Propylene Glycol; SBECD = Sulfobutylester beta cyclodextrin; MC = Methylcellulose; DMA = Dimethylaniline

Claims (19)

  1. A compound of Formula (A) or Formula (B)
    Figure imgb0051
    wherein
    R1 is H, (C1-C4)alkyl, (C1-C4)alkoxy, Cl, F, cyano, fluoro-substituted (C1-C2)alkyl, (C1-C4)alkyl-SO2-, or (C3-C6)cycloalkyl; and
    R2 is (C1-C4)alkyl, (C1-C4)alkoxy, (C2-C4)alkynyl, 3-oxetanyloxy, 3-tetrahydrofuranyloxy, Cl, F, cyano, fluoro-substituted (C1-C2)alkyl, (C1-C4)alkyl-SO2-, (C3-C6)cycloalkyl, or a (C5-C6)heterocycle having 1 or 2 heteroatoms each independently selected from N, O, or S.
  2. The compound of claim 1 wherein said compound is a compound of Formula (A).
  3. The compound of claims 1 or 2 wherein
    R1 is H, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, F, Cl, cyano, -CF3, cyclopropyl, or cyclobutyl; and
    R2 is methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, F, Cl, cyano, -CF3, -CF2CH3, ethynyl, 3-oxetanyloxy, 3-tetrahydrofuranyloxy, or cyclopropyl.
  4. The compound of claim 3 wherein
    R1 is H, methyl, ethyl, isopropyl, methoxy, ethoxy, F, Cl, cyano, -CF3, or cyclopropyl; and
    R2 is methyl, ethyl, isopropyl, methoxy, ethoxy, F, Cl, cyano, -CF3,
    -CF2CH3, ethynyl, 3-oxetanyloxy, 3-tetrahydrofuranyloxy, or cyclopropyl.
  5. The compound of claim 4 wherein
    R1 is H, methyl, ethyl, methoxy, ethoxy, F, Cl, cyano, -CF3, or cyclopropyl; and
    R2 is methyl, ethyl, methoxy, ethoxy, F, Cl, cyano, -CF3, -CF2CH3, ethynyl, 3-oxetanyloxy, 3-tetrahydrofuranyloxy, or cyclopropyl.
  6. The compound of claim 5 wherein
    R1 is methyl, ethyl, F, Cl, cyano, CF3, or cyclopropyl; and
    R2 is methoxy, or ethoxy.
  7. A compound selected from the group consisting of:
    (1S,2S,3S,4R,5S)-1-hydroxymethyl-5-[3-(4-methoxy-benzyl)-4-methyl-phenyl]-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol;
    (1 S,2S,3S,4R,5S)-5-[3-(4-ethoxy-benzyl)-4-methyl-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol;
    (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol;
    (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol;
    (1S,2S,3S,4R,5S)-5-[4-fluoro-3-(4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol;
    2-(4-methoxybenzyl)-4-((1S,2S,3S,4R,5S)-2,3,4-trihydroxy-1-(hydroxymethyl)-6,8-dioxa-bicyclo[3,2,1]oct-5-yl)benzonitrile;
    2-(4-ethoxybenzyl)-4-((1S,2S,3S,4R,5S)-2,3,4-trihydroxy-1-(hydroxymethyl)-6,8-dioxa-bicyclo[3,2,1]oct-5-yl)benzonitrile;
    (1 S,2S,3S,4R,5S)-5-[3-(4-ethoxy-benzyl)-4-fluoro-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol;
    (1S,2S,3S,4R,5S)-5-{4-fluoro-3-[4-(tetrahydro-furan-3-yloxy)-benzyl]-phenyl}-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol;
    (1S,2S, 3S,4R,5S)-5-[3-(4-chlorobenzyl)-4-fluorophenyl]-1-hydroxymethyl-6,8-dioxabicyclo[3.2.1]octane-2,3,4-triol;
    (1S,2S,3S,4R,5S)-5-{4-fluoro-3-[4-(oxetan-3-yloxy)-benzyl]-phenyl}-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; and
    (1S,2S,3S,4R,5S)-5-{4-chloro-3-[4-(oxetan-3-yloxy)-benzyl]-phenyl}-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol.
  8. A compound which is (1S,2S,3S,4R,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol.
  9. A compound selected from the group consisting of:
    (1S,2S,3S,4S,5S)-1-hydroxymethyl-5-[3-(4-methoxy-benzyl)-4-methyl-phenyl]-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol;
    (1 S,2S,3S,4S,5S)-5-[3-(4-ethoxy-benzyl)-4-methyl-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol;
    (1S,2S,3S,4S,5S)-5-[4-chloro-3-(4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol;
    (1S,2S,3S,4S,5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol;
    (1S,2S,3S,4S,5S)-5-[4-fluoro-3-(4-methoxy-benzyl)-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol;
    (1S,2S,3S,4S,5S)-5-[3-(4-ethoxy-benzyl)-4-fluoro-phenyl]-1-hydroxymethyl-6,8-dioxa-bicyclo[3.2.1]octane-2,3,4-triol; and
    (1S,2S,3S,4S,5S)-5-[3-(4-chlorobenzyl)-4-fluorophenyl]-1-hydroxymethyl-6,8-dioxabicyclo[3.2.1]octane-2,3,4-triol.
  10. A crystal comprising a compound having the formula (4A):
    Figure imgb0052
  11. The crystal of claim 10 further comprising L-proline or L-pyroglutamic acid.
  12. The crystal of claim 10 further comprising L-pyroglutamic acid and having one or more of the following:
    a) space group of P2(1)2(1)2(1) and unit cell parameters substantially equal to the following:
    a = 7.4907(10) Å α= 90°.
    b = 12.8626(15) Å β=90°.
    c = 28.029(4) Å γ= 90°;
    b) a powder x-ray diffraction pattern comprising 2-theta values of (CuKα radiation, wavelength of 1.54056Å) 6.4 ± 0.2, 16.7 ± 0.2, 17.4 ± 0.2 and 21.1 ± 0.2; or
    c) a solid state 13C NMR spectrum comprising peak positions at 16.5 ± 0.2, 131.1 ± 0.2, 158.7 ± 0.2, and 181.5 ± 0.2 ppm as determined on a 500MHz spectrometer relative to crystalline adamantine of 29.5 ppm.
  13. The crystal of claim 10 further comprising L-pyroglutamic acid wherein the crystal is a co-crystal comprising the compound of formula (4A) and L-pyroglutamic acid in a 1:1 stoichiometric ratio.
  14. The crystal of claim 10 further comprising L-pyroglutamic acid and having
    b) a powder x-ray diffraction pattern comprising a 2-theta value of (CuKα radiation, wavelength of 1.54056A) 6.4 ± 0.2; and
    d) a solid state 13C NMR spectrum comprising peak positions at 16.5 ± 0.2, 158.7 ± 0.2, and 181.5 ± 0.2 ppm as determined on a 500MHz spectrometer relative to crystalline adamantine of 29.5 ppm.
  15. The crystal of claim 10 further comprising L-proline and having one or more of the following:
    a) space group of C2 and unit cell parameters substantially equal to the following:
    a = 32.8399(16) Å α= 90°.
    b = 7.2457(4) Å β= 101.268(5)°.
    c = 11.8023(6) Å γ = 90°; or
    b) a powder x-ray diffraction pattern comprising 2-theta values of (CuKα radiation, wavelength of 1.54056A) 7.6 ± 0.2, 12.1 ± 0.2, 20.3 ± 0.2 and 28.8 ± 0.2.
  16. A pharmaceutical composition comprising (i) a compound of any of claims 1 through 9 or a crystal of any of claims 10-15; and (ii) a pharmaceutically acceptable excipient, diluent, or carrier.
  17. A compound of any of claims 1 through 9, a crystal of any of claims 10-15 or a pharmaceutical composition of claim 16 for use as a medicament.
  18. A compound of any of claims 1 through 9, a crystal of any of claims 10-15 or a pharmaceutical composition of claim 16 for use in the treatment of obesity and obesity-related disorders in animals.
  19. A compound of any of claims 1 through 9, a crystal of any of claims 10-15 or a pharmaceutical composition of claim 16 for use in treating or delaying the progression or onset of Type 2 diabetes and diabetes-related disorders in animals.
EP09786960A 2008-08-28 2009-08-17 Dioxa-bicyclo[3.2.1.]octane-2,3,4-triol derivatives Active EP2334687B9 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
PL09786960T PL2334687T3 (en) 2008-08-28 2009-08-17 Dioxa-bicyclo[3.2.1.]octane-2,3,4-triol derivatives
MEP-2011-45A ME01285A (en) 2008-08-28 2009-08-17 Dioxa-bicyclo[3.2.1.]octane-2,3,4-triol derivatives
CY20121100280T CY1112497T1 (en) 2008-08-28 2012-03-15 DIAXA-BICYCLE PRODUCERS [3.2.1] OCTAN-2,3,4-TRIOLIS
HUS1800031C HUS1800031I1 (en) 2008-08-28 2018-07-13 Dioxa-bicyclo[3.2.1.]octane-2,3,4-triol derivatives

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US9247008P 2008-08-28 2008-08-28
US22721209P 2009-07-21 2009-07-21
PCT/IB2009/053626 WO2010023594A1 (en) 2008-08-28 2009-08-17 Dioxa-bicyclo[3.2.1.]octane-2,3,4-triol derivatives

Publications (3)

Publication Number Publication Date
EP2334687A1 EP2334687A1 (en) 2011-06-22
EP2334687B1 EP2334687B1 (en) 2012-01-04
EP2334687B9 true EP2334687B9 (en) 2012-08-08

Family

ID=41208284

Family Applications (1)

Application Number Title Priority Date Filing Date
EP09786960A Active EP2334687B9 (en) 2008-08-28 2009-08-17 Dioxa-bicyclo[3.2.1.]octane-2,3,4-triol derivatives

Country Status (47)

Country Link
US (1) US8080580B2 (en)
EP (1) EP2334687B9 (en)
JP (1) JP4825322B1 (en)
KR (2) KR101446454B1 (en)
CN (2) CN103497199B (en)
AP (1) AP2728A (en)
AR (1) AR073138A1 (en)
AT (1) ATE540040T1 (en)
AU (1) AU2009286380B2 (en)
BR (1) BRPI0918841B8 (en)
CA (1) CA2733795C (en)
CL (1) CL2011000394A1 (en)
CO (1) CO6341636A2 (en)
CR (1) CR20110077A (en)
CY (2) CY1112497T1 (en)
DK (1) DK2334687T5 (en)
DO (1) DOP2011000058A (en)
EA (1) EA018492B1 (en)
EC (1) ECSP11010854A (en)
ES (1) ES2380408T3 (en)
FR (1) FR18C1036I2 (en)
GE (1) GEP20135803B (en)
HK (2) HK1156616A1 (en)
HN (1) HN2009001652A (en)
HR (1) HRP20120104T1 (en)
HU (1) HUS1800031I1 (en)
IL (2) IL211226A (en)
LT (1) LTC2334687I2 (en)
MA (1) MA32590B1 (en)
ME (1) ME01285A (en)
MX (1) MX2011002166A (en)
MY (1) MY155418A (en)
NI (1) NI201100043A (en)
NL (1) NL300943I2 (en)
NO (1) NO2018019I2 (en)
NZ (1) NZ591027A (en)
PA (1) PA8840801A1 (en)
PE (1) PE20110288A1 (en)
PL (1) PL2334687T3 (en)
PT (1) PT2334687E (en)
RS (1) RS52236B (en)
SI (1) SI2334687T1 (en)
SV (1) SV2011003842A (en)
TW (1) TWI387598B (en)
UY (1) UY32073A (en)
WO (1) WO2010023594A1 (en)
ZA (1) ZA201101341B (en)

Families Citing this family (112)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI523652B (en) 2008-07-15 2016-03-01 泰瑞克公司 Deuterated benzylbenzene derivatives and methods of use
SI2496583T1 (en) * 2009-11-02 2015-02-27 Pfizer Inc. Dioxa-bicycloš3.2.1ćoctane-2,3,4-triol derivatives
HUE025774T2 (en) 2010-03-19 2016-05-30 Pfizer 2,3 dihydro-1h-inden-1-yl-2,7-diazaspiro[3.6]nonane derivatives and their use as antagonists or inverse agonists of the ghrelin receptor
CN102372722A (en) * 2010-08-10 2012-03-14 江苏恒瑞医药股份有限公司 C-aryl glucoside derivative, preparation method thereof and application of C-aryl glucoside derivative in medicine
WO2012041898A1 (en) 2010-09-29 2012-04-05 Celon Pharma Sp. Z O.O. Combination of sglt2 inhibitor and a sugar compound for the treatment of diabetes
RU2540337C2 (en) 2010-10-29 2015-02-10 Пфайзер Инк. N1/N2-LACTAM ACETYL-CoA-CARBOXYLASE INHIBITORS
CN102643256B (en) * 2011-02-18 2014-12-24 上海璎黎科技有限公司 Arylglucoside compound and preparation method and application thereof
US20130345392A1 (en) 2011-03-04 2013-12-26 Pfizer Inc Edn3-like peptides and uses thereof
PL2699576T3 (en) 2011-04-22 2016-05-31 Pfizer Pyrazolospiroketone derivatives for use as acetyl-coa carboxylase inhibitors
WO2012163990A1 (en) 2011-06-03 2012-12-06 Boehringer Ingelheim International Gmbh Sglt-2 inhibitors for treating metabolic disorders in patients treated with neuroleptic agents
JP2014517032A (en) * 2011-06-13 2014-07-17 パナセア バイオテック リミテッド Novel SGLT inhibitor
TWI510491B (en) * 2011-06-30 2015-12-01 Shanghai Hengrui Pharm Co Ltd C-aryl glucoside derivatives, preparation process and pharmaceutical use thereof
JP2014520879A (en) 2011-07-15 2014-08-25 ファイザー・インク GPR119 regulator
US8927577B2 (en) 2011-07-22 2015-01-06 Pfizer Inc. Quinolinyl glucagon receptor modulators
EP2751116B1 (en) 2011-08-31 2016-10-12 Pfizer Inc Hexahydropyrano [3,4-d][1,3]thiazin-2-amine compounds
JP2014530186A (en) 2011-09-13 2014-11-17 パナセア バイオテック リミテッド Novel SGLT inhibitor
WO2013068439A1 (en) 2011-11-09 2013-05-16 Intervet International B.V. 4-amino-5-oxo-7,8-dihydropyrimido[5, 4 -f] [1, 4] oxazepine compounds as dgat1 inhibitors
UA108713C2 (en) 2011-11-11 2015-05-25 2-Thiopyrimidinones
MA37385B1 (en) 2012-04-06 2019-09-30 Pfizer Inc Etat De Delaware Diacylglycerol-acyltransferase 2 inhibitors
US8889730B2 (en) 2012-04-10 2014-11-18 Pfizer Inc. Indole and indazole compounds that activate AMPK
US9193751B2 (en) 2012-04-10 2015-11-24 Theracos, Inc. Process for the preparation of benzylbenzene SGLT2 inhibitors
ES2585262T3 (en) 2012-05-04 2016-10-04 Pfizer Inc Hexahydropyran [3,4-d] [1,3] thiazin-2-amine heterocyclic compounds substituted as inhibitors of PPA, BACE1 and BACE2
CN103570657A (en) * 2012-07-19 2014-02-12 天津药物研究院 Phenyl-glucoside derivative containing gem-dimethyl, preparation method and use thereof
US9145434B2 (en) 2012-07-26 2015-09-29 Boehringer Ingelheim International Gmbh Crystalline complex of 1-cyano-2-(4-cyclopropyl-benzyl)-4-(ss-d-glucopyranos-1-yl)-benzene, methods for its preparation and the use thereof for preparing medicaments
EP2897964A1 (en) 2012-09-20 2015-07-29 Pfizer Inc. Alkyl-substituted hexahydropyrano [3,4-d][1,3]thiazin-2-amine compounds
CN103772449B (en) * 2012-10-26 2017-12-26 上海阳帆医药科技有限公司 C aryl glucoside derivatives and preparation method thereof and purposes
WO2014086099A1 (en) * 2012-12-05 2014-06-12 广东东阳光药业有限公司 Glucopyranosyl derivative, preparation method and medical use thereof
CA2893256A1 (en) 2012-12-11 2014-06-19 Pfizer Inc. Hexahydropyrano [3,4-d][1,3]thiazin-2-amine compounds as inhibitors of bace1
JP5944593B2 (en) * 2012-12-17 2016-07-05 ティエンジン インスティテュート オブ ファーマシューティカル リサーチ Phenyl C-glucoside derivative having deoxyglucose structure, preparation method and use thereof
EP2935282A1 (en) 2012-12-19 2015-10-28 Pfizer Inc. CARBOCYCLIC- AND HETEROCYCLIC-SUBSTITUTED HEXAHYDROPYRANO[3,4-d][1,3]THIAZIN-2-AMINE COMPOUNDS
EP2956458B1 (en) 2013-02-13 2017-08-09 Pfizer Inc Heteroaryl-substituted hexahydropyrano[3,4-d][1,3]thiazin-2-amine compounds
US9233981B1 (en) 2013-02-15 2016-01-12 Pfizer Inc. Substituted phenyl hexahydropyrano[3,4-d][1,3]thiazin-2-amine compounds
EP3466431B1 (en) * 2013-03-14 2023-11-15 MSD International GmbH Crystalline forms and methods for preparing sglt2 inhibitors
CN104045513A (en) * 2013-03-14 2014-09-17 爱康药业有限公司 4-substituted-1-chloro-2-(4-fluorobenzyl)benzene, its preparation method and application as intermediate in preparation of anti-type II diabetes drugs
PT2981269T (en) 2013-04-04 2023-10-10 Boehringer Ingelheim Vetmedica Gmbh Treatment of metabolic disorders in equine animals
CN104619713B (en) * 2013-05-24 2017-06-16 四川海思科制药有限公司 Oxabicyclo derivative, preparation method and applications
WO2015027963A1 (en) * 2013-09-02 2015-03-05 四川海思科制药有限公司 Aromatic ring derivative, and pharmaceutical composition and use thereof
ES2605886T3 (en) * 2013-09-27 2017-03-16 Sunshine Lake Pharma Co., Ltd. Glucopiranosyl derivatives and their uses in medicine
WO2015052610A1 (en) 2013-10-09 2015-04-16 Pfizer Inc. Antagonists of prostaglandin ep3 receptor
CN105611920B (en) 2013-10-12 2021-07-16 泰拉科斯萨普有限责任公司 Preparation of hydroxy-diphenylmethane derivatives
CN111494357A (en) 2013-12-17 2020-08-07 勃林格殷格翰动物保健有限公司 Treatment of metabolic disorders in felines
MX2016009421A (en) 2014-01-23 2016-09-16 Boehringer Ingelheim Vetmedica Gmbh Treatment of metabolic disorders in canine animals.
AP2016009407A0 (en) 2014-03-17 2016-08-31 Pfizer Diacylglycerol acyltransferase 2 inhibitors for use in the treatment of metabolic and related disorders
HUE050095T2 (en) 2014-04-01 2020-11-30 Boehringer Ingelheim Vetmedica Gmbh Treatment of metabolic disorders in equine animals
SG11201608110WA (en) 2014-04-04 2016-10-28 Pfizer Bicyclic-fused heteroaryl or aryl compounds and their use as irak4 inhibitors
EA030085B1 (en) 2014-04-10 2018-06-29 Пфайзер Инк. 2-AMINO-6-METHYL-4,4a,5,6-TETRAHYDROPYRANO[3,4-d][1,3]THIAZIN-8a(8H)-YL-1,3-THIAZOL-4-YL AMIDES
SG11201608936UA (en) * 2014-05-19 2016-12-29 Pfizer Substituted-6,8-dioxabicyclo[3.2.1]octane-2,3-diol compounds as targeting agents of asgpr
CN104031098A (en) * 2014-06-21 2014-09-10 李友香 Hypoglycemic medicine
CN104017031A (en) * 2014-06-21 2014-09-03 李友香 Hypoglycemic drug and composition
EP3197429A1 (en) 2014-09-25 2017-08-02 Boehringer Ingelheim Vetmedica GmbH Combination treatment of sglt2 inhibitors and dopamine agonists for preventing metabolic disorders in equine animals
CN105461762B (en) * 2014-09-27 2018-12-04 广东东阳光药业有限公司 Glucopyranosyl derivatives and its application in medicine
CN105992769B (en) * 2014-09-30 2019-04-02 江苏恒瑞医药股份有限公司 A kind of L-PROLINE compound, its monohydrate and the crystal of white 2 inhibitor of sodium glucose co-transporter 2
WO2016077126A1 (en) 2014-11-10 2016-05-19 Merck Sharp & Dohme Corp. Sglt-2 inhibitors for treating metabolic disorders in patients with renal impairment or chronic kidney disease
WO2016092413A1 (en) 2014-12-10 2016-06-16 Pfizer Inc. Indole and indazole compounds that activate ampk
WO2016103097A1 (en) 2014-12-22 2016-06-30 Pfizer Inc. Antagonists of prostaglandin ep3 receptor
MX2017014128A (en) 2015-05-05 2018-03-15 Pfizer 2-thiopyrimidinones.
WO2016189463A1 (en) * 2015-05-25 2016-12-01 Sun Pharmaceutical Industries Limited Ertugliflozin co-crystals and process for their preparation
JP2018516254A (en) 2015-05-29 2018-06-21 ファイザー・インク Novel heterocyclic compounds as inhibitors of vanin 1 enzyme
EP3310784B1 (en) 2015-06-17 2020-10-07 Pfizer Inc Tricyclic compounds and their use as phosphodiesterase inhibitors.
WO2016203335A1 (en) 2015-06-18 2016-12-22 Pfizer Inc. Novel pyrido[2,3-b]pyrazinones as bet-family bromodomain inhibitors
BR112018002071A2 (en) 2015-08-13 2018-09-18 Pfizer fused bicyclic heteroaryl or aryl compounds
EP3341024A1 (en) 2015-08-27 2018-07-04 Boehringer Ingelheim Vetmedica GmbH Liquid pharmaceutical compositions comprising sglt-2 inhibitors
KR102029124B1 (en) 2015-08-27 2019-10-07 화이자 인코포레이티드 Bicyclic-Fused Heteroaryl or Aryl Compounds as IRAK4 Modulators
WO2017037567A1 (en) 2015-09-03 2017-03-09 Pfizer Inc. Regulators of frataxin
JP2018531924A (en) 2015-09-24 2018-11-01 ファイザー・インク Tetrahydropyrano [3,4-D] [1,3] oxazine derivatives and their use as BACE inhibitors
JP2018534251A (en) 2015-09-24 2018-11-22 ファイザー・インク N- [2- (3-Amino-2,5-dimethyl-1,1-dioxide-5,6-dihydro-2H-1,2,4-thiadiazin-5-yl) -1 useful as a BACE inhibitor , 3-Thiazol-4-yl] amide
WO2017051276A1 (en) 2015-09-24 2017-03-30 Pfizer Inc. N-[2-(2-amino-6,6-disubstituted-4, 4a, 5, 6-tetrahydropyrano [3,4-d][1,3] thiazin-8a (8h)-yl) -1, 3-thiazol-4-yl] amides
TN2018000198A1 (en) 2015-12-29 2019-10-04 Pfizer Substituted 3-azabicyclo[3.1.0]hexanes as ketohexokinase inhibitors
AU2017205545B2 (en) * 2016-01-04 2019-11-21 Je Il Pharmaceutical Co., Ltd. C-glucoside derivative containing fused phenyl ring or pharmaceutically acceptable salt thereof, process for preparing same, and pharmaceutical composition comprising same
CN105646603A (en) * 2016-03-01 2016-06-08 孙霖 Crystal form A of ertugliflozin and preparation method
CN105646604A (en) * 2016-03-01 2016-06-08 孙霖 Crystal form B of ertugliflozin and preparation method
EP3735975A1 (en) 2016-03-11 2020-11-11 Merck Sharp & Dohme Corp. Methods of treating or reducing the risk of cardiovascular events and related diseases using sglt-2 inhibitors
JP2019524716A (en) 2016-07-14 2019-09-05 ファイザー・インク Novel pyrimidinecarboxamides as inhibitors of vanin 1 enzyme
WO2018011721A1 (en) * 2016-07-15 2018-01-18 Granules India Limited Novel polymorphic forms of ((1s,2s,3s,4r,5s))-2,3,4-(tris-benzyloxy)-5-(4-chloro-3-(4-ethoxy-benzyl)phenyl)-6,8-dioxa-bicyclo[3.2.1]oct-1-yl-methanol
AR109179A1 (en) 2016-08-19 2018-11-07 Pfizer DIACILGLICEROL ACILTRANSFERASA 2 INHIBITORS
WO2018073154A1 (en) 2016-10-19 2018-04-26 Boehringer Ingelheim International Gmbh Combinations comprising an ssao/vap-1 inhibitor and a sglt2 inhibitor, uses thereof
CN107382952B (en) * 2017-07-13 2020-02-28 杭州科巢生物科技有限公司 Preparation method of eggliflozin and intermediate thereof
CN107898764A (en) * 2017-12-12 2018-04-13 威海贯标信息科技有限公司 A kind of net compositions of Ai Gelie
WO2019133445A1 (en) 2017-12-28 2019-07-04 Inception Ibd, Inc. Aminothiazoles as inhibitors of vanin-1
CN107857768A (en) * 2018-01-04 2018-03-30 威海贯标信息科技有限公司 A kind of net novel crystal forms of Ai Gelie
WO2019169988A1 (en) * 2018-03-06 2019-09-12 广东东阳光药业有限公司 Crystal forms of ertugliflozin and preparation method therefor
EP3781166A1 (en) 2018-04-17 2021-02-24 Boehringer Ingelheim International GmbH Pharmaceutical composition, methods for treating and uses thereof
WO2019202149A1 (en) 2018-04-19 2019-10-24 Université Catholique de Louvain Sglt2 inhibitors for the treatment of neutropenia
WO2020001812A1 (en) 2018-06-25 2020-01-02 Pharmathen S.A. A novel process for the preparation of sglt-2 inhibitors
WO2020026273A1 (en) * 2018-07-31 2020-02-06 Msn Laboratories Private Limited, R&D Center Solid forms of ertugliflozin free base and solid dispersions comprising ertugliflozin l-pyroglutamic acid.
WO2020039394A1 (en) 2018-08-24 2020-02-27 Novartis Ag New drug combinations
MX2021002428A (en) 2018-08-31 2023-01-02 Pfizer Combinations for treatment of nash/nafld and related diseases.
WO2020102575A1 (en) 2018-11-16 2020-05-22 Inception Ibd, Inc. Heterocyclic aminothiazoles and uses thereof
CA3140972A1 (en) 2019-05-20 2020-11-26 Pfizer Inc. Combinations comprising benzodioxol as glp-1r agonists for use in the treatment of nash/nafld and related diseases
TW202115086A (en) 2019-06-28 2021-04-16 美商輝瑞大藥廠 Bckdk inhibitors
WO2020261144A1 (en) 2019-06-28 2020-12-30 Pfizer Inc. 5-(thiophen-2-yl)-1h-tetrazole derivatives as bckdk inhibitors useful for treating various diseases
TW202116746A (en) * 2019-07-10 2021-05-01 大陸商廣東東陽光藥業有限公司 Glucopyranosyl derivative and application thereof
TWI771766B (en) 2019-10-04 2022-07-21 美商輝瑞股份有限公司 Diacylglycerol acyltransferase 2 inhibitor
JP7441946B2 (en) 2019-11-28 2024-03-01 ベーリンガー インゲルハイム フェトメディカ ゲーエムベーハー Use of SGLT-2 inhibitors in dry milk of non-human animals
CN113330017B (en) * 2019-12-19 2023-01-31 上海研健新药研发有限公司 Purification method and application of SGLTs inhibitor
CA3167531A1 (en) 2020-02-17 2021-08-26 Boehringer Ingelheim Vetmedica Gmbh Use of sglt-2 inhibitors for the prevention and/or treatment of cardiac diseases in felines
JP2022058085A (en) 2020-02-24 2022-04-11 ファイザー・インク Combination of inhibitors of diacylglycerol acyltransferase 2 and inhibitors of acetyl-coa carboxylase
JP2023523596A (en) 2020-04-22 2023-06-06 バイエル アクチェンゲゼルシャフト Combination of finerenone and SGLT2 inhibitors for treating and/or preventing cardiovascular and/or renal disease
CR20220632A (en) 2020-06-09 2023-01-23 Pfizer Spiro compounds as melanocortin 4 receptor antagonists and uses thereof
WO2021260498A1 (en) * 2020-06-22 2021-12-30 Aurobindo Pharma Limited An improved purification process for the preparation of ertugliflozin and ertugliflozin l-pyroglutamic acid co-crystal
CN114195748B (en) * 2020-09-17 2023-11-14 上海森辉医药有限公司 Preparation method of sodium-glucose cotransporter 2 inhibitor
WO2023006745A1 (en) 2021-07-28 2023-02-02 Boehringer Ingelheim Vetmedica Gmbh Use of sglt-2 inhibitors for the prevention and/or treatment of hypertension in non-human mammals
KR20240041966A (en) 2021-07-28 2024-04-01 베링거잉겔하임베트메디카게엠베하 Use of SGLT-2 inhibitors for the prevention and/or treatment of cardiac disease in non-human mammals other than cats, especially dogs
WO2023006747A1 (en) 2021-07-28 2023-02-02 Boehringer Ingelheim Vetmedica Gmbh Use of sglt-2 inhibitors for the prevention and/or treatment of renal diseases in non-human mammals
WO2023026180A1 (en) 2021-08-26 2023-03-02 Pfizer Inc. Amorphous form of (s)-2-(5-((3-ethoxypyridin-2-yl)oxy)pyridin-3-yl)-n-(tetrahydrofuran-3- yl)pyrimidine-5-carboxamide
KR20230060921A (en) 2021-10-28 2023-05-08 유니셀랩 주식회사 Novel ertugliflozin prodrug
WO2023100061A1 (en) 2021-12-01 2023-06-08 Pfizer Inc. 3-phenyl-1-benzothiophene-2-carboxylic acid derivatives as branched-chain alpha keto acid dehydrogenase kinase inhibitors for the treatment of diabetes, kidney diseases, nash and heart failure
WO2023105387A1 (en) 2021-12-06 2023-06-15 Pfizer Inc. Melanocortin 4 receptor antagonists and uses thereof
WO2023169456A1 (en) 2022-03-09 2023-09-14 Gasherbrum Bio , Inc. Heterocyclic glp-1 agonists
WO2023198140A1 (en) 2022-04-14 2023-10-19 Gasherbrum Bio, Inc. Heterocyclic glp-1 agonists
US20230381101A1 (en) 2022-05-25 2023-11-30 Boehringer Ingelheim Vetmedica Gmbh Aqueous pharmaceutical compositions comprising sglt-2 inhibitors
WO2024075051A1 (en) 2022-10-07 2024-04-11 Pfizer Inc. Hsd17b13 inhibitors and/or degraders

Family Cites Families (146)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US247705A (en) * 1881-09-27 William a
US560230A (en) * 1896-05-19 powers
US486299A (en) * 1892-11-15 John k
US838442A (en) * 1906-05-31 1906-12-11 Oscar Mietens Compressed-air railway-train brake.
FR1499439A (en) * 1966-09-16 1967-10-27 Electrochimie Soc New polyol and process for its preparation
JPS5373598A (en) * 1976-12-11 1978-06-30 Teikoku Chem Ind Corp Ltd Saccharide derivatives and process for thir preparation
JPS5911600B2 (en) * 1981-04-30 1984-03-16 理化学研究所 3-azido-1,6-anhydro-3-deoxy-2,4-di-O-benzyl-β-D-glucopyranose and its production method
NZ214774A (en) 1986-01-08 1989-10-27 Nz Scientific & Ind Res Herbicidal and/or plant growth regulatory compositions based on 1,6-anhydro-beta-hexopyranose derivatives and certain of these derivatives
US5041541A (en) 1988-05-05 1991-08-20 The Procter & Gamble Company Functional sugar substituted with reduced calories
CN1020944C (en) 1990-01-30 1993-05-26 阿图尔-费希尔股份公司费希尔厂 Fastening element
CA2079544A1 (en) 1991-10-04 1993-04-05 Adam Weislaw Mazur Cholesterol lowering compounds
DE4400464A1 (en) * 1994-01-11 1995-07-13 Bayer Ag Endoparasiticidal agents
DK0850948T3 (en) 1996-12-26 2002-07-29 Tanabe Seiyaku Co Propiophenone derivatives and process for their preparation
US6486299B1 (en) 1998-09-28 2002-11-26 Curagen Corporation Genes and proteins predictive and therapeutic for stroke, hypertension, diabetes and obesity
US6069238A (en) 1998-09-30 2000-05-30 Eli Lilly And Company Spirocyclic C-glycosides
PL203124B1 (en) 1999-08-31 2009-08-31 Kissei Pharmaceutical Glucopyranosyloxypyrazole derivatives, medicinal compositions containing the same and intermediates in the production thereof
US6515117B2 (en) 1999-10-12 2003-02-04 Bristol-Myers Squibb Company C-aryl glucoside SGLT2 inhibitors and method
PH12000002657B1 (en) 1999-10-12 2006-02-21 Bristol Myers Squibb Co C-aryl glucoside SGLT2 inhibitors
WO2001042469A1 (en) 1999-12-10 2001-06-14 Lexicon Genetics Incorporated Novel human transporter protein and polynucleotides encoding the same
US6627611B2 (en) 2000-02-02 2003-09-30 Kotobuki Pharmaceutical Co Ltd C-glycosides and preparation of thereof as antidiabetic agents
DE10006887A1 (en) 2000-02-16 2001-09-06 Hermann Koepsell Modulators of regulatory protein RS1, useful for treating diabetes or tumors and for increasing drug transport, control concentration of transport proteins in plasma membranes
DE10008128A1 (en) 2000-02-22 2001-08-23 Bayer Ag Endoparasiticide composition effective on topical administration, comprises solution of depsipeptide in solvent such as 1,2-isopropylidene-glycerol
BR0109323A (en) 2000-03-17 2002-12-24 Kissei Pharmaceutical Derivatives of gluco piranosiloxi benzyl benzene, medicinal compositions containing the same and intermediates for the preparation of derivatives
US6555519B2 (en) 2000-03-30 2003-04-29 Bristol-Myers Squibb Company O-glucosylated benzamide SGLT2 inhibitors and method
US6683056B2 (en) 2000-03-30 2004-01-27 Bristol-Myers Squibb Company O-aryl glucoside SGLT2 inhibitors and method
SI1329456T1 (en) 2000-09-29 2006-12-31 Kissei Pharmaceutical Glucopyranosyloxybenzylbenzene derivatives and medicinal compositions containing the same
EP1338603B1 (en) 2000-11-02 2010-01-20 Ajinomoto Co., Inc. Novel pyrazole derivatives and diabetes remedies containing the same
AU2002223127A1 (en) 2000-11-30 2002-06-11 Kissei Pharmaceutical Co., Ltd. Intellectual Property Glucopyranosyloxybenzyl benzene derivatives, medicinal compositions containing the same and intermediates in the production thereof
US20020081678A1 (en) 2000-12-11 2002-06-27 Gennady Merkulov Isolated nucleic acid molecules encoding human transporter proteins, and uses thereof
ES2326158T3 (en) 2000-12-28 2009-10-02 Kissei Pharmaceutical Co., Ltd. DERIVATIVES OF GLUCOPIRANOSILOXIPIRAZOL AND ITS USE AS MEDICINES.
EP1357186A4 (en) 2000-12-28 2005-01-26 Takeda Pharmaceutical Novel proteins and dnas thereof
TWI255817B (en) 2001-02-14 2006-06-01 Kissei Pharmaceutical Glucopyranosyloxybenzylbenzene derivatives and medicinal use thereof
DE60230591D1 (en) 2001-02-26 2009-02-12 Kissei Pharmaceutical GLYCOPYRANOSYLOXYPYRAZOLE DERIVATIVES AND THEIR MEDICAL USE
WO2002068440A1 (en) 2001-02-27 2002-09-06 Kissei Pharmaceutical Co., Ltd. Glycopyranosyloxypyrazole derivatives and medicinal use thereof
US6936590B2 (en) 2001-03-13 2005-08-30 Bristol Myers Squibb Company C-aryl glucoside SGLT2 inhibitors and method
WO2002081654A2 (en) 2001-03-27 2002-10-17 Pe Corporation (Ny) Isolated human transporter proteins, nucleic acid molecules encoding human transporter proteins, and uses thereof
WO2002080936A1 (en) 2001-04-04 2002-10-17 Ortho Mcneil Pharmaceutical, Inc. Combination therapy comprising glucose reabsorption inhibitors and ppar modulators
EP1392326B1 (en) 2001-04-04 2009-09-09 Ortho-McNeil-Janssen Pharmaceuticals, Inc. Combination therapy comprising glucose reabsorption inhibitors and retinoid-x receptor modulators
AU2002338643A1 (en) 2001-04-10 2002-10-28 Millennium Pharmaceuticals, Inc. 68723, sodium/glucose cotransporter family members and uses therefor
CA2444481A1 (en) 2001-04-11 2002-10-24 Bristol-Myers Squibb Company Amino acid complexes of c-aryl glucosides for treatment of diabetes and method
CA2672001A1 (en) 2001-04-27 2002-11-07 Ajinomoto Co., Inc. N-substituted pyrazole-o-glycoside derivatives and therapeutic agent for diabetes containing the same
EP1400529A4 (en) 2001-05-30 2007-12-19 Kissei Pharmaceutical Glucopyranosyloxypyrazole derivative, medicinal composition containing the same, medicinal use thereof, and intermediate therefor
JP4399254B2 (en) 2001-06-20 2010-01-13 キッセイ薬品工業株式会社 Nitrogen-containing heterocyclic derivative, pharmaceutical composition containing the same, pharmaceutical use thereof and production intermediate thereof
JP4115105B2 (en) 2001-07-02 2008-07-09 協和醗酵工業株式会社 Pyrazole derivative
US7082091B2 (en) * 2001-07-31 2006-07-25 Ricoh Company, Ltd. Information reproducing method judging a multivalued level of a present cell by referring to judged multivalued levels of a preceding cell and an ensuing cell
EP1489089A4 (en) 2002-03-22 2009-10-28 Kissei Pharmaceutical Crystals of glucopyranosyloxybenzyl benzene derivative
US20070059356A1 (en) 2002-05-31 2007-03-15 Almarsson Oern Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
DE10225844A1 (en) 2002-06-04 2003-12-18 Lang Florian sgk and nedd as diagnostic and therapeutic targets
DE10231370B4 (en) 2002-07-11 2006-04-06 Sanofi-Aventis Deutschland Gmbh Thiophene glycoside derivatives, medicaments containing these compounds and methods of making these medicaments
TWI254635B (en) 2002-08-05 2006-05-11 Yamanouchi Pharma Co Ltd Azulene derivative and salt thereof
CA2494179C (en) 2002-08-08 2012-04-24 Kissei Pharmaceutical Co., Ltd. Pyrazole derivative, medicinal composition containing the same, medicinal use thereof, and intermediate for production thereof
DE10237085A1 (en) 2002-08-09 2004-02-19 Bioagency Ag Treatment or prevention of parasitic helminth infections, especially filariasis, in humans or animals, using oxyamino-substituted phosphonic or phosphinic acid compounds
BR0310006A (en) 2002-08-09 2005-02-15 Taisho Pharmaceutical Co Ltd Aryl 5-thio-beta-d-glycopyranoside derivatives and diabetes therapeutic agents containing them
JP2004137245A (en) 2002-08-23 2004-05-13 Kissei Pharmaceut Co Ltd Pyrazole derivative, pharmaceutical composition containing the same, its pharmaceutical use and production intermediate
AU2003275713A1 (en) 2002-10-29 2004-05-25 Takeda Pharmaceutical Company Limited Use of sglt homolog
CN1744916A (en) 2002-12-04 2006-03-08 橘生药品工业株式会社 Preventive or remedy for diseases caused by hyperglycemia
JP4651934B2 (en) 2002-12-04 2011-03-16 キッセイ薬品工業株式会社 Benzylphenol derivative, pharmaceutical composition containing it, and pharmaceutical use thereof
DE10258007B4 (en) 2002-12-12 2006-02-09 Sanofi-Aventis Deutschland Gmbh Aromatic fluoroglycoside derivatives, medicaments containing these compounds and methods for the preparation of these medicaments
DE10258008B4 (en) 2002-12-12 2006-02-02 Sanofi-Aventis Deutschland Gmbh Heterocyclic fluoroglycoside derivatives, medicaments containing these compounds and methods of making these medicaments
AU2003289440A1 (en) 2002-12-25 2004-07-22 Kissei Pharmaceutical Co., Ltd. Nitrogen-containing heterocycic derivatives, medicinal compositions containing the same and medicinal use thereof
BR0317929A (en) 2003-01-03 2006-04-11 Bristol Myers Squibb Co methods of producing c-aryl glycoside sglt2 inhibitors
CN1771334A (en) 2003-03-07 2006-05-10 施瑞修德制药公司 Method for determining susceptibility of tumor to treatment with anti-neoplastic agent
US7202350B2 (en) 2003-03-14 2007-04-10 Astellas Pharma Inc. C-glycoside derivatives and salts thereof
JP2004300102A (en) 2003-03-31 2004-10-28 Kissei Pharmaceut Co Ltd Condensed heterocyclic derivative, pharmaceutical composition containing the same and its pharmaceutical application
EP1609799A4 (en) 2003-04-01 2008-10-29 Taisho Pharmaceutical Co Ltd Heteroaryl 5-thio-beta-d-glucopyranoside derivatives and remedies for diabetes containing the same
US7439232B2 (en) 2003-04-01 2008-10-21 Taisho Pharmaceutical Co., Ltd. Heteroaryl 5-thio-β-D-glucopyranoside derivatives and therapeutic agents for diabetes containing the same
AU2003902263A0 (en) 2003-05-12 2003-05-29 Fujisawa Pharmaceutical Co., Ltd. Monosaccharide compounds
JPWO2004106352A1 (en) 2003-05-29 2006-07-20 大正製薬株式会社 Method for producing aldohexopyranose intermediate
JP2004359630A (en) 2003-06-06 2004-12-24 Yamanouchi Pharmaceut Co Ltd Difluorodiphenylmethane derivative and its salt
EP1637539B1 (en) 2003-06-20 2012-01-18 Kissei Pharmaceutical Co., Ltd. Pyrazole derivative, drug composition containing the same and production intermediate therefor
UA86042C2 (en) 2003-08-01 2009-03-25 Янссен Фармацевтика Н.В. Substituted indazole-o-glucosides
AR045173A1 (en) 2003-08-01 2005-10-19 Tanabe Seiyaku Co COMPOUNDS THAT HAVE INHIBITORY ACTIVITY AGAINST TRANSPORTER OF GLUCOSE DEPENDENT ON SODIUM
US7375090B2 (en) 2003-08-26 2008-05-20 Boehringer Ingelheim International Gmbh Glucopyranosyloxy-pyrazoles, pharmaceutical compositions containing these compounds, the use thereof and processed for the preparation thereof
US7371732B2 (en) 2003-12-22 2008-05-13 Boehringer Ingelheim International Gmbh Glucopyranosyloxy-substituted aromatic compounds, medicaments containing such compounds, their use and process for their manufacture
JP2005247834A (en) 2004-02-04 2005-09-15 Taisho Pharmaceut Co Ltd Activity inhibitor of sodium-dependent glucose cotransporter 2
AU2005219776B2 (en) 2004-03-04 2010-07-08 Kissei Pharmaceutical Co., Ltd. Nitrogenous fused-ring derivatives, medicinal compositions containing the derivatives, and use thereof as drugs
US7732596B2 (en) 2004-03-04 2010-06-08 Kissei Pharmaceutical Co., Ltd. Fused heterocycle derivative, medicinal composition containing the same, and medicinal use thereof
BRPI0508259A (en) 2004-03-04 2007-07-31 Kissei Pharmaceutical fused heterocyclic derivative, medicinal composition containing same, and medicinal use thereof
EA011158B1 (en) 2004-03-16 2009-02-27 Бёрингер Ингельхайм Интернациональ Гмбх Glucopyranosyl-substituted benzol derivatives, drugs containing said compounds, the use thereof and method for the production thereof
DE102004012676A1 (en) * 2004-03-16 2005-10-06 Boehringer Ingelheim Pharma Gmbh & Co. Kg New glucopyranosyl-substituted benzene derivatives are sodium-dependent glucose cotransporter inhibitors, useful for treating e.g. metabolic disorders (type 1 and type 2 diabetes mellitus or metabolic acidosis)
WO2005095373A1 (en) 2004-03-31 2005-10-13 Kissei Pharmaceutical Co., Ltd. Naphthalene derivative, medicinal composition containing the same, and medicinal use thereof
WO2005095372A1 (en) 2004-03-31 2005-10-13 Kissei Pharmaceutical Co., Ltd. Naphthalene derivative, medicinal composition containing the same, and medicinal use thereof
EP1731524A4 (en) 2004-03-31 2009-05-20 Kissei Pharmaceutical Phenol derivative, medicinal composition containing the same, and medicinal use thereof
DE102004028241B4 (en) 2004-06-11 2007-09-13 Sanofi-Aventis Deutschland Gmbh New fluoroglycoside derivatives of pyrazoles, medicines containing these compounds and manufacture of these medicines
US7393836B2 (en) 2004-07-06 2008-07-01 Boehringer Ingelheim International Gmbh D-xylopyranosyl-substituted phenyl derivatives, medicaments containing such compounds, their use and process for their manufacture
JPWO2006006496A1 (en) 2004-07-08 2008-04-24 アステラス製薬株式会社 Method for producing azulene derivatives and synthetic intermediates thereof
DE102004034690A1 (en) 2004-07-17 2006-02-02 Boehringer Ingelheim Pharma Gmbh & Co. Kg Methylidene-D-xylopyranosyl and oxo-D-xylopyranosyl-substituted phenyls, medicaments containing these compounds, their use and processes for their preparation
JP5010918B2 (en) 2004-07-21 2012-08-29 キッセイ薬品工業株式会社 Inhibitors of disease progression caused by abnormal accumulation of liver fat
TW200606129A (en) 2004-07-26 2006-02-16 Chugai Pharmaceutical Co Ltd Novel cyclohexane derivative, its prodrug, its salt and diabetic therapeutic agent containing the same
WO2006010557A1 (en) 2004-07-27 2006-02-02 Boehringer Ingelheim International Gmbh D-glucopyranosyl phenyl-substituted cyclene, medicaments containing these compounds, their use, and method for the production thereof
WO2006018150A1 (en) 2004-08-11 2006-02-23 Boehringer Ingelheim International Gmbh D-xylopyranosyl-phenyl-substituited cyclene, medicaments containing said compounds, use thereof and method for the production thereof
AR051446A1 (en) 2004-09-23 2007-01-17 Bristol Myers Squibb Co C-ARYL GLUCOSIDS AS SELECTIVE INHIBITORS OF GLUCOSE CONVEYORS (SGLT2)
JP2006117651A (en) 2004-09-27 2006-05-11 Taisho Pharmaceut Co Ltd Sglt2 activity inhibitor
MX2007003785A (en) 2004-09-29 2007-07-12 Kissei Pharmaceutical 1-( ??-d-glycopyranosyl)-3-substituted nitrogenous heterocyclic compound, medicinal composition containing the same, and medicinal use thereof.
DE102004048388A1 (en) 2004-10-01 2006-04-06 Boehringer Ingelheim Pharma Gmbh & Co. Kg D-pyranosyl-substituted phenyls, pharmaceutical compositions containing them, their use and processes for their preparation
WO2006054629A1 (en) 2004-11-18 2006-05-26 Kissei Pharmaceutical Co., Ltd. 1-SUBSTITUTED-3-β-D-GLUCOPYRANOSYLATED NITROGENOUS HETERO- CYCLIC COMPOUNDS AND MEDICINES CONTAINING THE SAME
JP2008007405A (en) 2004-12-07 2008-01-17 Takeda Chem Ind Ltd Carboxamide derivative
US7687469B2 (en) 2004-12-16 2010-03-30 Boehringer Ingelheim International Gmbh Glucopyranosyl-substituted benzene derivatives, medicaments containing such compounds, their use and process for their manufacture
TW200637839A (en) 2005-01-07 2006-11-01 Taisho Pharmaceutical Co Ltd 1-thio-d-glucitol derivatives
TW200637869A (en) 2005-01-28 2006-11-01 Chugai Pharmaceutical Co Ltd The spiroketal derivatives and the use as therapeutical agent for diabetes of the same
AR053329A1 (en) 2005-01-31 2007-05-02 Tanabe Seiyaku Co INDOL DERIVATIVES USEFUL AS INHIBITORS OF GLUCOSE CONVEYORS DEPENDENT ON SODIUM (SGLT)
CA2597269A1 (en) 2005-02-15 2006-08-24 Kissei Pharmaceutical Co., Ltd. 1-substituted-7-(beta-d-glycopyranosyloxy)(aza)indole compound and pharmaceutical containing the same
ATE445608T1 (en) 2005-02-23 2009-10-15 Boehringer Ingelheim Int GLUCOPYRANOSYL-SUBSTITUTED ((HETERO)ARYLETHYNYL-BENZYL)-BENZENE DERIVATIVES AND THEIR USE AS INHIBITORS OF THE SODIUM-DEPENDENT GLUCOSE CO-TRANSPORTER TYPE 2 (SGLT2)
WO2006108584A2 (en) 2005-04-15 2006-10-19 Cenix Bioscience Gmbh Human marker genes and agents for cardiovascular disorders and artherosclerosis
WO2006108842A1 (en) 2005-04-15 2006-10-19 Boehringer Ingelheim International Gmbh Glucopyranosyl-substituted (heteroaryloxy-benzyl)-benzene derivatives as sglt inhibitors
JPWO2006115137A1 (en) 2005-04-22 2008-12-18 キッセイ薬品工業株式会社 2-Aminobenzimidazole derivatives and their pharmaceutical use
WO2006119038A1 (en) 2005-04-29 2006-11-09 Naturegen, Inc. Compositions and methods for controlling glucose uptake
US7723309B2 (en) 2005-05-03 2010-05-25 Boehringer Ingelheim International Gmbh Crystalline forms of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((R)-tetrahydrofuran-3-yloxy)-benzyl]-benzene, a method for its preparation and the use thereof for preparing medicaments
UA91546C2 (en) 2005-05-03 2010-08-10 Бьорінгер Інгельхайм Інтернаціональ Гмбх Crystalline form of 1-chloro-4-(я-d-glucopyranos-1-yl)-2-[4-((s)-tetrahydrofuran-3-yloxy)-benzyl]-benzene, a method for its preparation and the use thereof for preparing medicaments
US7772191B2 (en) 2005-05-10 2010-08-10 Boehringer Ingelheim International Gmbh Processes for preparing of glucopyranosyl-substituted benzyl-benzene derivatives and intermediates therein
WO2007000445A1 (en) 2005-06-29 2007-01-04 Boehringer Ingelheim International Gmbh Glucopyranosyl-substituted benzyl-benzene derivatives, medicaments containing such compounds, their use and process for their manufacture
TW200726755A (en) 2005-07-07 2007-07-16 Astellas Pharma Inc A crystalline choline salt of an azulene derivative
JP5128474B2 (en) 2005-07-27 2013-01-23 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Glucopyranosyl-substituted ((hetero) cycloalkylethynyl-benzyl) -benzene derivatives, agents containing the compounds, their use and methods for their production
WO2007019526A2 (en) 2005-08-05 2007-02-15 Naturegen, Inc. Compositions and methods for controlling glucose and lipid uptake from foods
WO2007025943A2 (en) 2005-08-30 2007-03-08 Boehringer Ingelheim International Gmbh Glucopyranosyl-substituted benzyl-benzene derivatives, medicaments containing such compounds, their use and process for their manufacture
US8507450B2 (en) 2005-09-08 2013-08-13 Boehringer Ingelheim International Gmbh Crystalline forms of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-ethynyl-benzyl)-benzene, methods for its preparation and the use thereof for preparing medicaments
US20070099237A1 (en) 2005-10-31 2007-05-03 The Regents Of The University Of Michigan Reaction co-crystallization of molecular complexes or co-crystals
TWI370818B (en) 2006-04-05 2012-08-21 Astellas Pharma Inc Cocrystal of c-glycoside derivative and l-proline
JP2009167104A (en) 2006-05-02 2009-07-30 Taisho Pharmaceutical Co Ltd Phenyl 5-thio glycoside compound
JP2009167103A (en) 2006-05-02 2009-07-30 Taisho Pharmaceutical Co Ltd Pyrazolyl 5-thioglycoside compound
PE20080697A1 (en) 2006-05-03 2008-08-05 Boehringer Ingelheim Int BENZONITRILE DERIVATIVES SUBSTITUTED WITH GLUCOPYRANOSIL, PHARMACEUTICAL COMPOSITIONS CONTAINING COMPOUNDS OF THIS TYPE, THEIR USE AND PROCEDURE FOR THEIR MANUFACTURE
JP5230613B2 (en) 2006-05-23 2013-07-10 テラコス・インコーポレイテッド Glucose transporter inhibitor and method of use thereof
DE102006028862A1 (en) 2006-06-23 2007-12-27 Merck Patent Gmbh 3-amino-imidazo [1,2-a] pyridine
US7919598B2 (en) 2006-06-28 2011-04-05 Bristol-Myers Squibb Company Crystal structures of SGLT2 inhibitors and processes for preparing same
US8115017B2 (en) 2006-06-29 2012-02-14 Taisho Pharmaceutical Co., Ltd C-phenyl 1-thioglucitol compound
TWI432446B (en) 2006-07-27 2014-04-01 Chugai Pharmaceutical Co Ltd Fused ring spiroketal derivative and use thereof as anti-diabetic drug
TWI418556B (en) 2006-07-27 2013-12-11 Mitsubishi Tanabe Pharma Corp Indole derivatives
JP2008050353A (en) 2006-07-27 2008-03-06 Mitsubishi Tanabe Pharma Corp Pharmaceutical composition
TWI403516B (en) * 2006-07-27 2013-08-01 Chugai Pharmaceutical Co Ltd To replace spirocyclic alcohol derivatives, and its use as a therapeutic agent for diabetes
US20080027014A1 (en) 2006-07-28 2008-01-31 Tanabe Seiyaku Co., Ltd. Novel SGLT inhibitors
TW200817424A (en) 2006-08-04 2008-04-16 Daiichi Sankyo Co Ltd Benzylphenyl glucopyranoside derivatives
CN101121148B (en) 2006-08-08 2010-05-12 中国科学院大连化学物理研究所 Direct forming method of fluidized reaction catalyst containing molecular sieve
JP5231416B2 (en) * 2006-08-09 2013-07-10 ダウ グローバル テクノロジーズ エルエルシー Multi-segment expandable polymer composition with controllable expansion direction
JP5384343B2 (en) 2006-08-15 2014-01-08 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Glucopyranosyl-substituted cyclopropylbenzene derivatives, pharmaceutical compositions containing such compounds, their use as SGLT inhibitors and methods for their preparation
US7858587B2 (en) 2006-09-21 2010-12-28 Boehringer Ingelheim International Gmbh Glucopyranosyl-substituted difluorobenzyl-benzene derivates, medicaments containing such compounds, their use and process for their manufacture
EP2072522A4 (en) 2006-10-13 2010-01-06 Chugai Pharmaceutical Co Ltd Thioglucose spiroketal derivative and use thereof as therapeutic agent for diabetes
DE102006048728A1 (en) 2006-10-16 2008-04-17 Merck Patent Gmbh 3-amino-imidazo {1,2-a] pyridine
CA2667550A1 (en) 2006-10-27 2008-05-02 Boehringer Ingelheim International Gmbh Crystalline form of 4-(.beta.-d-glucopyranos-1-yl)-1-methyl-2-[4-((s)-tetrahydrofuran-3-yloxy)-benzyl]-benzene, a method for its preparation and the use thereof for preparing medicaments
EP2079753A1 (en) 2006-11-06 2009-07-22 Boehringer Ingelheim International GmbH Glucopyranosyl-substituted benzyl-benzonitrile derivatives, medicaments containing such compounds, their use and process for their manufacture
WO2008115574A1 (en) 2007-03-21 2008-09-25 Reliant Pharmaceuticals, Inc. Cb1 antagonist and a dyslipidemic agent and/or metabolic regulator, and methods of making and using same
TW200904454A (en) 2007-03-22 2009-02-01 Bristol Myers Squibb Co Methods for treating obesity employing an SGLT2 inhibitor and compositions thereof
KR101663324B1 (en) 2007-07-26 2016-10-06 렉시컨 파마슈티컬스 인코퍼레이티드 Methods and compounds useful for the preparation of sodium glucose co-transporter 2 inhibitors
CL2008002425A1 (en) 2007-08-16 2009-09-11 Boehringer Ingelheim Int Pharmaceutical composition comprising an inhibitor of sglt2 and 1- (4-methyl-quinazolin-2-yl) methyl-3-methyl-7 - (- 2-butin-1-yl) -8- (3- (r) -amino- Piperidin-1yl) -xanthine, an iv dpp inhibitor and its use for the treatment of obesity and type 1 and 2 diabetes and complications thereof.
PL2187742T3 (en) 2007-08-23 2018-06-29 Theracos Sub, Llc (2s,3r,4r,5s,6r)-2-(4-chloro-3-benzylphenyl)-6-(hydroxymethyl)tetrahydro-2h-pyran-3,4,5-triol derivatives for use in the treatment of diabetes
CL2008003653A1 (en) 2008-01-17 2010-03-05 Mitsubishi Tanabe Pharma Corp Use of a glucopyranosyl-derived sglt inhibitor and a selected dppiv inhibitor to treat diabetes; and pharmaceutical composition.
CN101503399B (en) * 2008-02-04 2012-06-27 白鹭医药技术(上海)有限公司 C-aryl glucoside SGLT2 inhibitor

Also Published As

Publication number Publication date
CL2011000394A1 (en) 2011-06-17
AP2011005616A0 (en) 2011-04-30
BRPI0918841A2 (en) 2015-12-08
BRPI0918841B1 (en) 2019-10-08
ES2380408T3 (en) 2012-05-11
AR073138A1 (en) 2010-10-13
CY2018024I2 (en) 2019-11-27
JP2012500842A (en) 2012-01-12
MX2011002166A (en) 2011-04-07
TW201014863A (en) 2010-04-16
KR20130116078A (en) 2013-10-22
DK2334687T5 (en) 2012-09-10
NO2018019I2 (en) 2019-02-19
MY155418A (en) 2015-10-15
CR20110077A (en) 2011-03-09
FR18C1036I1 (en) 2018-10-05
FR18C1036I2 (en) 2020-07-24
HK1156616A1 (en) 2012-06-15
CA2733795A1 (en) 2010-03-04
AU2009286380A8 (en) 2011-03-17
ATE540040T1 (en) 2012-01-15
AU2009286380A1 (en) 2010-03-04
CN102149717B (en) 2014-05-14
KR20110045093A (en) 2011-05-03
DK2334687T3 (en) 2012-04-16
MA32590B1 (en) 2011-08-01
CO6341636A2 (en) 2011-11-21
HN2009001652A (en) 2012-02-13
CN103497199B (en) 2015-04-29
HK1193606A1 (en) 2014-09-26
KR101338540B1 (en) 2013-12-06
ZA201101341B (en) 2011-11-30
EA018492B1 (en) 2013-08-30
PL2334687T3 (en) 2012-05-31
SV2011003842A (en) 2011-03-23
JP4825322B1 (en) 2011-11-30
HRP20120104T1 (en) 2012-02-29
UY32073A (en) 2010-03-26
PE20110288A1 (en) 2011-05-26
US8080580B2 (en) 2011-12-20
LTPA2018510I1 (en) 2018-09-25
BRPI0918841B8 (en) 2021-05-25
TWI387598B (en) 2013-03-01
WO2010023594A1 (en) 2010-03-04
PT2334687E (en) 2012-04-13
PA8840801A1 (en) 2010-04-21
NZ591027A (en) 2012-11-30
EA201100266A1 (en) 2011-10-31
IL236804B (en) 2019-06-30
HUS1800031I1 (en) 2018-08-28
CY2018024I1 (en) 2019-11-27
ME01285A (en) 2013-06-20
IL211226A0 (en) 2011-04-28
LTC2334687I2 (en) 2019-09-10
NI201100043A (en) 2011-08-05
US20100056618A1 (en) 2010-03-04
ES2380408T9 (en) 2013-02-22
AP2728A (en) 2013-08-31
DOP2011000058A (en) 2011-03-15
EP2334687A1 (en) 2011-06-22
CA2733795C (en) 2013-12-31
RS52236B (en) 2012-10-31
GEP20135803B (en) 2013-04-10
IL236804A0 (en) 2015-03-31
AU2009286380B2 (en) 2011-09-15
EP2334687B1 (en) 2012-01-04
CN103497199A (en) 2014-01-08
SI2334687T1 (en) 2012-03-30
IL211226A (en) 2015-02-26
CY1112497T1 (en) 2015-12-09
NL300943I2 (en) 2018-08-07
ECSP11010854A (en) 2011-03-31
CN102149717A (en) 2011-08-10
NO2018019I1 (en) 2018-06-26
KR101446454B1 (en) 2014-10-06

Similar Documents

Publication Publication Date Title
EP2334687B9 (en) Dioxa-bicyclo[3.2.1.]octane-2,3,4-triol derivatives
EP2496583B1 (en) Dioxa-bicyclo[3.2.1]octane-2,3,4-triol derivatives
US10285973B2 (en) SGLT-2 inhibitors for treating metabolic disorders in patients with renal impairment or chronic kidney disease

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20110328

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

AX Request for extension of the european patent

Extension state: AL BA RS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

AX Request for extension of the european patent

Extension state: AL BA RS

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: E. BLUM & CO. AG PATENT- UND MARKENANWAELTE VSP

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 540040

Country of ref document: AT

Kind code of ref document: T

Effective date: 20120115

REG Reference to a national code

Ref country code: HR

Ref legal event code: TUEP

Ref document number: P20120104

Country of ref document: HR

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: HR

Ref legal event code: T1PR

Ref document number: P20120104

Country of ref document: HR

REG Reference to a national code

Ref country code: RO

Ref legal event code: EPE

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602009004618

Country of ref document: DE

Effective date: 20120308

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20120402

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Ref country code: EE

Ref legal event code: FG4A

Ref document number: E006325

Country of ref document: EE

Effective date: 20120224

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20120400265

Country of ref document: GR

Effective date: 20120322

REG Reference to a national code

Ref country code: NO

Ref legal event code: T2

Effective date: 20120104

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2380408

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20120511

REG Reference to a national code

Ref country code: PL

Ref legal event code: T3

REG Reference to a national code

Ref country code: SK

Ref legal event code: T3

Ref document number: E 11422

Country of ref document: SK

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E012997

Country of ref document: HU

REG Reference to a national code

Ref country code: DK

Ref legal event code: T5

RIN2 Information on inventor provided after grant (corrected)

Inventor name: COLLMAN, BENJAMIN, MICAH

Inventor name: MASCITTI, VINCENT

REG Reference to a national code

Ref country code: SK

Ref legal event code: TE4A

Ref document number: E 11422

Country of ref document: SK

Owner name: E 11422 2334687, 4.1.2012 COLLMAN BENJAMIN MIC, US

Effective date: 20090817

Ref country code: SK

Ref legal event code: T4

Ref document number: E 11422

Country of ref document: SK

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20121005

REG Reference to a national code

Ref country code: EE

Ref legal event code: LD4A

Ref document number: E006325

Country of ref document: EE

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602009004618

Country of ref document: DE

Effective date: 20121005

REG Reference to a national code

Ref country code: PL

Ref legal event code: RECP

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20120831

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120104

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 8

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 9

REG Reference to a national code

Ref country code: DE

Ref legal event code: R065

Ref document number: 602009004618

Country of ref document: DE

Free format text: PRODUCT NAME: ERTUGLIFLOZIN, EINSCHLIESSLICH KRISTALL UMFASSEND ERTUGLIFLOZIN UND ERTUGLIFLOZIN UND L-PYROGLUTAMINSAEURE ALS CO-KRISTALL; REGISTRATION NO/DATE: EU/1/18/1267 20180321

Spc suppl protection certif: 122018000070

Filing date: 20180626

Expiry date: 20290818

Extension date: 20330323

Ref country code: DE

Ref legal event code: R065

Ref document number: 602009004618

Country of ref document: DE

Free format text: PRODUCT NAME: ERTUGLIFLOZIN, GEGEBENENFALLS ALS EINE KRISTALLFORM, BEVORZUGT ALS EIN CO-KRISTALL MIT L-PYROGLUTAMINSAEURE, UND INSBESONDERE ALS ERTUGLIFLOZIN-L-PYROGLUTAMINSAEURE; REGISTRATION NO/DATE: EU/1/18/1267 20180321

Spc suppl protection certif: 122018000070

Filing date: 20180626

Expiry date: 20290818

REG Reference to a national code

Ref country code: NO

Ref legal event code: SPCF

Free format text: PRODUCT NAME: ERTUGLIFLOZIN, EVENTUELT SOM; REG. NO/DATE: EU/1/18/1267 20180409

Spc suppl protection certif: 2018019

Filing date: 20180626

REG Reference to a national code

Ref country code: FI

Ref legal event code: SPCF

Spc suppl protection certif: C20180024

Ref country code: NL

Ref legal event code: SPCF

Free format text: PRODUCT NAME: ERTUGLIFLOZINE, DESGEWENST IN KRISTALVORM, MET NAME ALS CO-KRISTAL MET L-PYROGLUTAMINEZUUR, MET NAME ERTUGLIFLOZIN L-PYROGLUTAMINE ZUUR; REGISTRATION NO/DATE: EU/1/18/1267 20180323

Spc suppl protection certif: 300943

Filing date: 20180627

Expiry date: 20290816

Extension date: 20330322

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 10

REG Reference to a national code

Ref country code: IT

Ref legal event code: SPCF

Ref document number: 502012902028239

Country of ref document: IT

Free format text: PRODUCT NAME: ERTUGLIFLOZIN(STEGLATRO); AUTHORISATION NUMBER(S) AND DATE(S): EU/1/18/1267, 20180323

Spc suppl protection certif: 132018000000441

REG Reference to a national code

Ref country code: GB

Ref legal event code: CTFF

Free format text: PRODUCT NAME: ERTUGLIFOZIN, OPTIONALLY AS A CRYSTAL FORM, PARTICULARLY AS A CO-CRYSTAL WITH L-PYROGLUTAMIC ACID, AND SPECIFICALLY AS ERTUGLIFOZIN L-PYROGLUTAMIC ACID; REGISTERED: UK EU/1/18/1267 20180321

Spc suppl protection certif: SPC/GB18/026

Filing date: 20180704

REG Reference to a national code

Ref country code: SE

Ref legal event code: SPCF

Free format text: PRODUCT NAME: ERTUGLIFLOZIN,OPTIONALLY AS A CRYSTAL FORM, PARTICULARLY AS A CO-CRYSTAL WITH L-PYROGUTAMIC ACID, AND SPECIFICALLY AS ERTUGLIFLOZIN L-PYROGLUTAMIC ACID; REG. NO/DATE: EU/1/18/1267 20180323

Spc suppl protection certif: 1890026-6

REG Reference to a national code

Ref country code: NL

Ref legal event code: SPCG

Free format text: PRODUCT NAME: ERTUGLIFLOZINE, DESGEWENST IN KRISTALVORM, MET NAME ALS CO-KRISTAL MET L-PYROGLUTAMINEZUUR, MET NAME ERTUGLIFLOZIN L-PYROGLUTAMINE ZUUR; REGISTRATION NO/DATE: EU/1/18/1267 20180323

Spc suppl protection certif: 300943

Filing date: 20180627

Expiry date: 20290816

Extension date: 20330322

Effective date: 20180807

Ref country code: IE

Ref legal event code: SPCF

Free format text: PRODUCT NAME: ERTUGLIFLOZIN, OPTIONALLY AS A CRYSTAL FORM, PARTICULARLY AS A CO-CRYSTAL WITH L-PYROGLUTAMIC ACID, AND SPECIFICALLY AS ERTUGLIFLOZIN L-PYROGLUTAMIC ACID; REGISTRATION NO/DATE: EU/1/18/1267/001 EU/1/18/1267/012 20180321

Spc suppl protection certif: 2018/028

Filing date: 20180710

REG Reference to a national code

Ref country code: IT

Ref legal event code: SPCG

Ref document number: 502012902028239

Country of ref document: IT

Free format text: PRODUCT NAME: ERTUGLIFLOZIN(STEGLATRO); AUTHORISATION NUMBER(S) AND DATE(S): EU/1/18/1267, 20180323

Spc suppl protection certif: 132018000000441

Extension date: 20330323

Ref country code: LU

Ref legal event code: SPCF

Free format text: PRODUCT NAME: ERTUGLIFLOZINE, EVENTUELLEMENT SOUS FORME CRISTALLINE, EN PARTICULIER EN TANT QUE CO-CRISTAL AVEC L'ACIDE L-PYROGLUTAMIQUE, ET PLUS SPECIFIQUEMENT EN TANT QU'ACIDE ERTUGLIFLOZINE L-PYROGLUTAMIQUE; AUTHORISATION NUMBER AND DATE: EU/1/18/1267 20180323

Spc suppl protection certif: LUC00079

Filing date: 20180627

Expiry date: 20290817

Extension date: 20330323

REG Reference to a national code

Ref country code: FR

Ref legal event code: CP

Free format text: PRODUCT NAME: ERTUGLIFLOZINE,OPTIONNELLEMENT SOUS FORME CRISTALLINE,EN PARTICULIER EN TANT QUE CO-CRISTAL AVEC L'ACIDE L-PYROGLUTANIQUE,ET PARTICULIEREMENT ERTUGLIFOZINE ACIDE L-PYROGLUTANIQUE.; REGISTRATION NO/DATE: EU/1/18/1267 20180323

Spc suppl protection certif: 18C1036

Filing date: 20180824

Ref country code: FR

Ref legal event code: CP

Free format text: PRODUCT NAME: ERTUGLIFLOZINE,OPTIONNELLEMENT SOUS FORME CRISTALLINE,EN PARTICULIER EN TANT QUE CO-CRISTAL AVEC L'ACIDE L-PYROGLUTANIQUE,ET PARTICULIEREMENT ERTUGLIFOZINE ACIDE L-PYROGLUTANIQUE.; REGISTRATION NO/DATE: EU/1/18/1267 20180321

Spc suppl protection certif: 18C1036

Filing date: 20180824

REG Reference to a national code

Ref country code: HU

Ref legal event code: AA1S

Ref document number: E012997

Country of ref document: HU

Spc suppl protection certif: S1800031

Filing date: 20180713

REG Reference to a national code

Ref country code: BE

Ref legal event code: SPCF

Free format text: PRODUCT NAME: ERTUGLIFLOZINE, DESGEWENST IN KRISTALVORM, MET NAME ALS CO-KRISTAL MET L-PYROGLUTAMINEZUUR, EN SPECIFIEK ALS ERTUGLIFLOZIN L-PYROGLUTAMINE ZUUR; AUTHORISATION NUMBER AND DATE: EU/1/18/1267 20180323

Spc suppl protection certif: 2018C/027

Filing date: 20180627

Expiry date: 20290817

Extension date: 20330323

REG Reference to a national code

Ref country code: AT

Ref legal event code: SPCF

Ref document number: 540040

Country of ref document: AT

Kind code of ref document: T

Free format text: PRODUCT NAME: ERTUGLIFLOZIN, GEGEBENENFALLS ALS KRISTALLFORM, INSBESONDERE ALS CO-KRISTALL MIT L-PYROGLUTAMINSAEURE, UND INSBESONDERE ALS ERTUGLIFLOZIN L-PYROGLUTAMINSAEURE; REGISTRATION NO/DATE: EU/1/18/1267 (MITTEILUNG) 20180323

Spc suppl protection certif: 30/2018

Filing date: 20180726

Effective date: 20180915

REG Reference to a national code

Ref country code: EE

Ref legal event code: AA1Y

Ref document number: E006325

Country of ref document: EE

Free format text: PRODUCT NAME: ERTUGLIFLOSIIN;REG NO/DATE: EU/1/18/1267 23.03.2018

Spc suppl protection certif: C20180023

Filing date: 20180824

REG Reference to a national code

Ref country code: LT

Ref legal event code: SPCF

Free format text: PRODUCT NAME: ERTUGLIFLOZINAS, PASIRINKTINAI KAIP KRISTALINE FORMA, YPAC KAIP KO-KRISTALAS SU L-PIROGLUTAMO RUGSTIMI, IR YPAC KAIP ERTUGLIFLOZINO L-PIROGLUTAMO RUGSTIS; REGISTRATION NO/DATE: EU/1/18/1267 20180321

Spc suppl protection certif: PA2018510

Filing date: 20180821

Expiry date: 20290817

REG Reference to a national code

Ref country code: DK

Ref legal event code: CTFF

Free format text: PRODUCT NAME: ERTUGLIFLOZIN, EVENTUELT SOM EN KRYSTALFORM, SAERLIGT SOM ET CO-KRYSTAL MED L-PYROGLUTAMINSYRE OG SPECIFIKT SOM ERTUGLIFLOZIN-L-PYROGLUTAMINSYRE; NAT. REG. NO/DATE: EU/1/18/1267/001-012 20180323; FIRST REG. NO/DATE: EU EU/1/18/1267/001/012 20180323

Spc suppl protection certif: CA 2018 00025

Filing date: 20180628

Expiry date: 20290817

Extension date: 20330323

REG Reference to a national code

Ref country code: LU

Ref legal event code: SPCG

Free format text: PRODUCT NAME: ERTUGLIFLOZINE, EVENTUELLEMENT SOUS FORME CRISTALLINE, EN PARTICULIER EN TANT QUE CO-CRISTAL AVEC L'ACIDE L-PYROGLUTAMIQUE, ET PLUS SPECIFIQUEMENT EN TANT QU'ACIDE ERTUGLIFLOZINE L-PYROGLUTAMIQUE; AUTHORISATION NUMBER AND DATE: EU/1/18/1267 20180323

Spc suppl protection certif: LUC00079

Filing date: 20180627

Expiry date: 20290817

Extension date: 20330323

Effective date: 20180907

REG Reference to a national code

Ref country code: AT

Ref legal event code: SPCG

Ref document number: 540040

Country of ref document: AT

Kind code of ref document: T

Free format text: PRODUCT NAME: ERTUGLIFLOZIN, GEGEBENENFALLS ALS KRISTALLFORM, INSBESONDERE ALS CO-KRISTALL MIT L-PYROGLUTAMINSAEURE, UND INSBESONDERE ALS ERTUGLIFLOZIN L-PYROGLUTAMINSAEURE; REGISTRATION NO/DATE: EU/1/18/1267 (MITTEILUNG) 20180323

Spc suppl protection certif: 30/2018

Filing date: 20180726

Expiry date: 20290817

Extension date: 20330323

Effective date: 20181115

REG Reference to a national code

Ref country code: ES

Ref legal event code: SPCF

Free format text: PRODUCT NAME: ERTUGLIFLOZINA, OPICIONALMENTE EN FORMA CRISTALINA, PARTICULARMENTE COMO UN CO-CRISTAL CON ACIDO L-PIROGLUTAMICO, Y ESPECIFICAMENTE COMO ACIDO L-PIROGLUTAMICO DE ERTUGLIFLOZINA; NATIONAL AUTHORISATION NUMBER: EU/1/18/1267; DATE OF AUTHORISATION: 20180321; NUMBER OF FIRST AUTHORISATION IN EUROPEAN ECONOMIC AREA (EEA): EU/1/18/1267; DATE OF FIRST AUTHORISATION IN EEA: 20180321

Spc suppl protection certif: C201830047

Effective date: 20180905

REG Reference to a national code

Ref country code: SK

Ref legal event code: SPCF

Free format text: PRODUCT NAME: ERTUGLIFLOZIN VO VSETKYCH FORMACH CHRANENYCH ZAKLADNYM PATENTOM; REGISTRATION NO/DATE: EU/1/18/1267 20180323

Spc suppl protection certif: 18-2018

Filing date: 20180917

REG Reference to a national code

Ref country code: CH

Ref legal event code: SPCF

Free format text: PRODUCT NAME: ERTUGLIFLOZIN; REGISTRATION NO/DATE: SWISSMEDIC AUTHORISATION 66577 18.10.2018

Filing date: 20181212

REG Reference to a national code

Ref country code: NO

Ref legal event code: SPCG

Free format text: PRODUCT NAME: ERTUGLIFLOZIN, EVENTUELT SOM; REG. NO/DATE: EU/1/18/1267 20180409

Spc suppl protection certif: 2018019

Filing date: 20180626

REG Reference to a national code

Ref country code: EE

Ref legal event code: FG1Y

Ref document number: E006325

Country of ref document: EE

Free format text: PRODUCT NAME: ERTUGLIFLOSIIN;REG NO/DATE: EU/1/18/1267 23.03.2018

Spc suppl protection certif: C20180023 00266

Filing date: 20180824

Extension date: 20330323

REG Reference to a national code

Ref country code: ES

Ref legal event code: SPCG

Free format text: PRODUCT NAME: ERTUGLIFLOZINA, OPICIONALMENTE EN FORMA CRISTALINA, PARTICULARMENTE COMO UN CO-CRISTAL CON ACIDO L-PIROGLUTAMICO, Y ESPECIFICAMENTE COMO ACIDO L-PIROGLUTAMICO DE ERTUGLIFLOZINA; NATIONAL AUTHORISATION NUMBER: EU/1/18/1267; DATE OF AUTHORISATION: 20180321; NUMBER OF FIRST AUTHORISATION IN EUROPEAN ECONOMIC AREA (EEA): EU/1/18/1267; DATE OF FIRST AUTHORISATION IN EEA: 20180321

Spc suppl protection certif: C201830047

Extension date: 20330323

Effective date: 20190611

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20120104

Country of ref document: HR

Payment date: 20190807

Year of fee payment: 11

REG Reference to a national code

Ref country code: LT

Ref legal event code: SPCG

Free format text: PRODUCT NAME: ERTUGLIFLOZINAS, PASIRINKTINAI KAIP KRISTALINE FORMA, YPAC KAIP KO-KRISTALAS SU L-PIROGLUTAMO RUGSTIMI, IR YPAC KAIP ERTUGLIFLOZINO L-PIROGLUTAMO RUGSTIS; REGISTRATION NO/DATE: EU/1/18/1267 20180321

Spc suppl protection certif: PA2018510,C2334687

Filing date: 20180821

Expiry date: 20290817

Extension date: 20330323

REG Reference to a national code

Ref country code: DK

Ref legal event code: CTFG

Free format text: PRODUCT NAME: ERTUGLIFLOZIN, EVENTUELT SOM EN KRYSTALFORM, SAERLIGT SOM ET CO-KRYSTAL MED L-PYROGLUTAMINSYRE OG SPECIFIKT SOM ERTUGLIFLOZIN-L-PYROGLUTAMINSYRE; NAT. REG. NO/DATE: EU/1/18/1267/001-012 20180323; FIRST REG. NO/DATE: EU EU/1/18/1267/001/012 20180323

Spc suppl protection certif: CR 2018 00025

Filing date: 20180628

Expiry date: 20290817

Extension date: 20330323

REG Reference to a national code

Ref country code: SE

Ref legal event code: SPCG

Free format text: PRODUCT NAME: ERTUGLIFLOZIN,OPTIONALLY IN THE FORM OF ERTUGLIFOZIN L- PYROGLUTAMIC ACID; REG. NO/DATE: EU/1/18/1267 20180323

Spc suppl protection certif: 1890026-6

Expiry date: 20290818

Extension date: 20330322

REG Reference to a national code

Ref country code: IE

Ref legal event code: SPCG

Free format text: PRODUCT NAME: ERTUGLIFLOZIN, OPTIONALLY AS A CRYSTAL FORM, PARTICULARLY AS A CO-CRYSTAL WITH L-PYROGLUTAMIC ACID, AND SPECIFICALLY AS ERTUGLIFLOZIN L-PYROGLUTAMIC ACID; REGISTRATION NO/DATE: EU/1/18/1267/001 EU/1/18/1267/012 20180321

Spc suppl protection certif: 2018/028

Extension date: 20330322

Effective date: 20191217

REG Reference to a national code

Ref country code: CH

Ref legal event code: SPCG

Free format text: PRODUCT NAME: ERTUGLIFLOZIN; REGISTRATION NO/DATE: SWISSMEDIC AUTHORISATION 66577 18.10.2018

Spc suppl protection certif: C02334687/01

Filing date: 20181212

Extension date: 20331017

REG Reference to a national code

Ref country code: DE

Ref legal event code: R067

Ref document number: 602009004618

Country of ref document: DE

Free format text: PRODUCT NAME: ERTUGLIFLOZIN, EINSCHLIESSLICH KRISTALL UMFASSEND ERTUGLIFLOZIN UND ERTUGLIFLOZIN UND L-PYROGLUTAMINSAEURE ALS CO-KRISTALL; REGISTRATION NO/DATE: EU/1/18/1267 20180321

Spc suppl protection certif: 122018000070

Filing date: 20180626

Expiry date: 20290818

Extension date: 20330323

REG Reference to a national code

Ref country code: DE

Ref legal event code: R069

Ref document number: 602009004618

Country of ref document: DE

Free format text: PRODUCT NAME: ERTUGLIFLOZIN, EINSCHLIESSLICH KRISTALL UMFASSEND ERTUGLIFLOZIN UND ERTUGLIFLOZIN UND L-PYROGLUTAMINSAEURE ALS CO-KRISTALL; REGISTRATION NO/DATE: EU/1/18/1267 20180321

Spc suppl protection certif: 122018000070

Filing date: 20180626

Expiry date: 20290818

Extension date: 20330323

REG Reference to a national code

Ref country code: HU

Ref legal event code: FG4S

Ref document number: E012997

Country of ref document: HU

Spc suppl protection certif: S1800031

Filing date: 20180713

REG Reference to a national code

Ref country code: FR

Ref legal event code: CT

Free format text: PRODUCT NAME: ERTUGLIFLOZINE,OPTIONNELLEMENT SOUS FORME CRISTALLINE,EN PARTICULIER EN TANT QUE CO-CRISTAL AVEC L'ACIDE L-PYROGLUTANIQUE,ET PARTICULIEREMENT ERTUGLIFOZINE ACIDE L-PYROGLUTANIQUE.; REGISTRATION NO/DATE: EU/1/18/1267 20180323

Spc suppl protection certif: 18C1036

Filing date: 20180824

Extension date: 20330323

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20120104

Country of ref document: HR

Payment date: 20200806

Year of fee payment: 12

REG Reference to a national code

Ref country code: SK

Ref legal event code: SPCG

Free format text: PRODUCT NAME: ERTUGLIFLOZIN VO VSETKYCH FORMACH CHRANENYCH ZAKLADNYM PATENTOM; REGISTRATION NO/DATE: EU/1/18/1267 20180323

Spc suppl protection certif: 392 18-2018

Filing date: 20180917

Extension date: 20330323

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20120104

Country of ref document: HR

Payment date: 20210811

Year of fee payment: 13

REG Reference to a national code

Ref country code: GB

Ref legal event code: CTFG

Free format text: PRODUCT NAME: ERTUGLIFOZIN, OPTIONALLY AS A CRYSTAL FORM, PARTICULARLY AS A CO-CRYSTAL WITH L-PYROGLUTAMIC ACID, AND SPECIFICALLY AS ERTUGLIFOZIN L-PYROGLUTAMIC ACID; REGISTERED: UK EU/1/18/1267/001(NI) 20180321; UK EU/1/18/1267/002(NI) 20180321; UK EU/1/18/1267/003(NI) 20180321; UK EU/1/18/1267/004(NI) 20180321; UK EU/1/18/1267/005(NI) 20180321; UK EU/1/18/1267/006(NI) 20180321; UK PLGB 53095/0064 20180321; UK PLGB 53095/0065 20180321; UK EU/1/18/1267/007(NI) 20180321; UK EU/1/18/1267/008(NI) 20180321; UK EU/1/18/1267/009(NI) 20180321; UK EU/1/18/1267/010(NI) 20180321; UK EU/1/18/1267/011(NI) 20180321; UK EU/1/18/1267/012(NI) 20180321

Spc suppl protection certif: SPC/GB18/026

Filing date: 20180704

Extension date: 20330322

Effective date: 20211206

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20120104

Country of ref document: HR

Payment date: 20220810

Year of fee payment: 14

REG Reference to a national code

Ref country code: FI

Ref legal event code: SPCG

Spc suppl protection certif: 716

Extension date: 20330323

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MK

Payment date: 20220713

Year of fee payment: 14

REG Reference to a national code

Ref country code: CH

Ref legal event code: SPCP

Owner name: PFIZER INC., US

Free format text: FORMER OWNER: PFIZER INC., US

Spc suppl protection certif: C02334687/01

REG Reference to a national code

Ref country code: LU

Ref legal event code: SPCT

Owner name: PFIZER INC.; US

Spc suppl protection certif: LUC00079

Effective date: 20230314

Ref country code: HR

Ref legal event code: PNAN

Ref document number: P20120104

Country of ref document: HR

Owner name: PFIZER INC., US

Ref country code: LU

Ref legal event code: HC

Owner name: PFIZER INC.; US

Free format text: FORMER OWNER: PFIZER INC.

Effective date: 20230314

REG Reference to a national code

Ref country code: LT

Ref legal event code: SPCT

Free format text: PRODUCT NAME: ERTUGLIFLOZINAS, PASIRINKTINAI KAIP KRISTALINE FORMA, YPAC KAIP KO-KRISTALAS SU L-PIROGLUTAMO RUGSTIMI, IR YPAC KAIP ERTUGLIFLOZINO L-PIROGLUTAMO RUGSTIS; REGISTRATION NO/DATE: EU/1/18/1267 20180321

Spc suppl protection certif: PA2018510,C2334687

Filing date: 20180821

Expiry date: 20290817

Extension date: 20330323

REG Reference to a national code

Ref country code: HU

Ref legal event code: HC9C

Owner name: PFIZER INC., US

Free format text: FORMER OWNER(S): PFIZER INC., US

REG Reference to a national code

Ref country code: BE

Ref legal event code: PD

Owner name: PFIZER INC.; US

Free format text: DETAILS ASSIGNMENT: CHANGE OF OWNER(S), OTHER; FORMER OWNER NAME: PFIZER INC.

Effective date: 20230316

REG Reference to a national code

Ref country code: SK

Ref legal event code: TC4A

Ref document number: E 11422

Country of ref document: SK

Owner name: PFIZER INC., NEW YORK, NY, US

Effective date: 20230419

REG Reference to a national code

Ref country code: DE

Ref legal event code: R081

Ref document number: 602009004618

Country of ref document: DE

Owner name: PFIZER INC. (N.D.GES.D.STAATES DELAWARE), NEW , US

Free format text: FORMER OWNER: PFIZER INC., 10017 NEW YORK, N.Y., US

REG Reference to a national code

Ref country code: NO

Ref legal event code: CHAD

Owner name: PFIZER INC., US

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230320

REG Reference to a national code

Ref country code: SI

Ref legal event code: SP73

Owner name: PFIZER INC.; US

Effective date: 20230523

REG Reference to a national code

Ref country code: FR

Ref legal event code: SPCC

Owner name: PFIZER INC., US

Free format text: PRODUCT NAME: ERTUGLIFLOZINE,OPTIONNELLEMENT SOUS FORME CRISTALLINE,EN PARTICULIER EN TANT QUE CO-CRISTAL AVEC L'ACIDE L-PYROGLUTANIQUE,ET PARTICULIEREMENT ERTUGLIFOZINE ACIDE L-PYROGLUTANIQUE.; REGISTRATION NO/DATE: EU/1/18/1267 20180323

Spc suppl protection certif: 18C1036

Filing date: 20180824

Extension date: 20330323

Ref country code: EE

Ref legal event code: SPCT

Owner name: PFIZER INC., NEW YORK, NY 10001 2192, US

Free format text: PREVIOUS OWNER: PFIZER, INC., NEW YORK, NY 10017, US

Spc suppl protection certif: 00266

Ref country code: EE

Ref legal event code: HE1A

Ref document number: E006325

Country of ref document: EE

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PT

Payment date: 20230628

Year of fee payment: 15

Ref country code: DK

Payment date: 20230627

Year of fee payment: 15

Ref country code: BG

Payment date: 20230629

Year of fee payment: 15

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20120104

Country of ref document: HR

Payment date: 20230721

Year of fee payment: 15

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PL

Payment date: 20230619

Year of fee payment: 15

Ref country code: NL

Payment date: 20230720

Year of fee payment: 15

Ref country code: LU

Payment date: 20230713

Year of fee payment: 15

Ref country code: IS

Payment date: 20230621

Year of fee payment: 15

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: TR

Payment date: 20230809

Year of fee payment: 15

Ref country code: RO

Payment date: 20230809

Year of fee payment: 15

Ref country code: NO

Payment date: 20230726

Year of fee payment: 15

Ref country code: MT

Payment date: 20230615

Year of fee payment: 15

Ref country code: IT

Payment date: 20230810

Year of fee payment: 15

Ref country code: IE

Payment date: 20230726

Year of fee payment: 15

Ref country code: GB

Payment date: 20230706

Year of fee payment: 15

Ref country code: FI

Payment date: 20230727

Year of fee payment: 15

Ref country code: ES

Payment date: 20230906

Year of fee payment: 15

Ref country code: EE

Payment date: 20230712

Year of fee payment: 15

Ref country code: CZ

Payment date: 20230717

Year of fee payment: 15

Ref country code: CY

Payment date: 20230712

Year of fee payment: 15

Ref country code: CH

Payment date: 20230901

Year of fee payment: 15

Ref country code: AT

Payment date: 20230726

Year of fee payment: 15

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SK

Payment date: 20230711

Year of fee payment: 15

Ref country code: SI

Payment date: 20230712

Year of fee payment: 15

Ref country code: SE

Payment date: 20230808

Year of fee payment: 15

Ref country code: HU

Payment date: 20230803

Year of fee payment: 15

Ref country code: HR

Payment date: 20230721

Year of fee payment: 15

Ref country code: GR

Payment date: 20230726

Year of fee payment: 15

Ref country code: FR

Payment date: 20230710

Year of fee payment: 15

Ref country code: DE

Payment date: 20230711

Year of fee payment: 15

Ref country code: BE

Payment date: 20230712

Year of fee payment: 15

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: LV

Payment date: 20230712

Year of fee payment: 15

Ref country code: LT

Payment date: 20230804

Year of fee payment: 15

REG Reference to a national code

Ref country code: CH

Ref legal event code: SPCN

Spc suppl protection certif: C02334687/01

Representative=s name: E. BLUM AND CO. AG, CH

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MK

Payment date: 20230712

Year of fee payment: 15